WO2020067439A1 - Sheeting method for cardiomyocytes - Google Patents

Sheeting method for cardiomyocytes Download PDF

Info

Publication number
WO2020067439A1
WO2020067439A1 PCT/JP2019/038192 JP2019038192W WO2020067439A1 WO 2020067439 A1 WO2020067439 A1 WO 2020067439A1 JP 2019038192 W JP2019038192 W JP 2019038192W WO 2020067439 A1 WO2020067439 A1 WO 2020067439A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
culture
graft
cardiomyocytes
Prior art date
Application number
PCT/JP2019/038192
Other languages
French (fr)
Japanese (ja)
Inventor
芳樹 澤
繁 宮川
賢二 大山
文哉 大橋
Original Assignee
国立大学法人大阪大学
テルモ株式会社
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 国立大学法人大阪大学, テルモ株式会社 filed Critical 国立大学法人大阪大学
Priority to JP2020549444A priority Critical patent/JP7256818B2/en
Publication of WO2020067439A1 publication Critical patent/WO2020067439A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/34Muscles; Smooth muscle cells; Heart; Cardiac stem cells; Myoblasts; Myocytes; Cardiomyocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P41/00Drugs used in surgical methods, e.g. surgery adjuvants for preventing adhesion or for vitreum substitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/48Reproductive organs
    • A61K35/54Ovaries; Ova; Ovules; Embryos; Foetal cells; Germ cells
    • A61K35/545Embryonic stem cells; Pluripotent stem cells; Induced pluripotent stem cells; Uncharacterised stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M3/00Tissue, human, animal or plant cell, or virus culture apparatus

Definitions

  • the present invention relates to a method, a graft manufactured using the method, a method for treating a disease using the graft, and the like.
  • Non-Patent Document 1 As a source of cardiomyocytes used for preparing such grafts, cardiomyocytes derived from pluripotent stem cells such as embryonic stem cells (ES cells) and induced pluripotent stem cells (iPS cells) have recently been receiving attention. Production of sheet-shaped cell cultures containing such pluripotent stem cell-derived cardiomyocytes and treatment experiments on animals have been attempted (Non-patent Documents 2 and 3). However, the development of sheet-like cell cultures containing cardiomyocytes derived from pluripotent stem cells has only just begun, and there are still many unknowns regarding their functional properties and factors affecting them.
  • ES cells embryonic stem cells
  • iPS cells induced pluripotent stem cells
  • the present disclosure relates to a method for producing a transplant such as a high-quality sheet-shaped cell culture, which includes iPS cell-derived differentiation-inducing cells, particularly cardiomyocytes, while maintaining the function of the cells at a high level, and using the method. It is an object of the present invention to provide a graft manufactured by the method, a method for treating a disease using the graft, and the like.
  • a graft containing cardiomyocytes to be used for transplantation When preparing a graft containing cardiomyocytes to be used for transplantation, it must be prepared in a xeno-free environment. In addition, when using fetal bovine serum (FBS) or a serum-free medium, a problem such as insufficient pulsation of cardiomyocytes has been found.
  • FBS fetal bovine serum
  • the present inventors have attempted to prepare a sheet-shaped cell culture for living body transplantation using iPS cell-derived cardiomyocytes having a sufficient function even if prepared in a xenofree environment, Using a platelet (lysate), a new finding was found that a stronger pulsation could be observed in a shorter time than when serum was used. Based on such findings, further research has been carried out to demonstrate that it is possible to produce high-quality transplants that can withstand clinical applications in the preparation of transplants containing various somatic cells differentiated from iPS cells. As a result, the present invention has been completed.
  • the present invention relates to the following: [1] A method for producing a transplant containing cells induced to differentiate from pluripotent stem cells; (A) a step of inoculating a cell population containing the cells on a culture substrate, and (b) a step of explanting and culturing the inoculated cell population with an explant forming medium containing a platelet lysate;
  • the above method comprising: [2] The method of [1], wherein the pluripotent stem cells are iPS cells. [3] The method of [1] or [2], wherein the transplant is a sheet-shaped cell culture.
  • a method for accelerating the onset of pulsation of cardiomyocytes in a graft containing cardiomyocytes differentiated from iPS cells, wherein the cell population containing cardiomyocytes is transplanted with a graft-forming medium containing platelet lysate comprising culturing a piece.
  • a transplant such as a sheet-shaped cell culture, having a higher quality than before can be produced with high efficiency from a cell population including differentiation-inducing cells, particularly cardiomyocytes, that have been induced to differentiate from iPS cells.
  • differentiation-inducing cells particularly cardiomyocytes
  • cardiomyocytes differentiated from iPS cells it is possible to prepare grafts retaining desired properties at a high level. It is possible to provide a graft which is very suitable for transplantation.
  • FIG. 1 is a table showing a comparison of the results when sheet culture was performed using the serum-free medium and the PL-containing medium in Example 2 respectively.
  • the PL-containing medium was used, sheets were obtained in all lots after two days of culture, but when the serum-free medium was used, sheets could not be formed due to breakage in half of the lots.
  • FIG. 2 is a graph comparing the state of pulsation of a sheet-shaped cardiomyocyte culture prepared using an FBS-containing medium and using a PL-containing medium. A stronger pulsation was observed earlier in the medium prepared with the PL-containing medium than in the medium prepared with the FBS-containing medium.
  • the present disclosure provides a method for producing an implant comprising cells induced to differentiate from pluripotent stem cells; (A) inoculating a cell population containing the cells on a culture substrate, and (b) explanting and culturing the inoculated cell population in a medium containing platelet lysate;
  • the method relates to:
  • the term “graft” refers to a structure for transplantation into a living body, and particularly refers to a structure for transplantation containing cells as a component.
  • the so-called suspension state in which at least one state in which cells are adhered to each other in a transplant to form a certain shape as a whole, and each and every cell is present separately, is referred to as the present disclosure.
  • the implant is an implantable structure that does not include structures other than cells and cell-derived substances (eg, a scaffold).
  • Examples of the graft in the present disclosure include, but are not limited to, a sheet-shaped cell culture, a spheroid, a cell aggregate, a cell suspension, a cell suspension containing fibrin gel, and a cell using a nanofiber. Cultures and the like are preferable, and a sheet cell culture or a spheroid is preferable, and a sheet cell culture is more preferable.
  • the “sheet-shaped cell culture” refers to a cell in which cells are connected to each other to form a sheet.
  • spheroid refers to a cell in which cells are connected to each other to form a substantially spherical shape.
  • the cells may be connected to each other directly (including via a cellular element such as an adhesion molecule) and / or via an intermediary substance.
  • the intervening substance is not particularly limited as long as it is a substance capable of at least physically (mechanically) connecting cells, and examples thereof include an extracellular matrix.
  • the intervening substance is preferably derived from cells, particularly from cells constituting a sheet-shaped cell culture or spheroid.
  • the sheet-shaped cell culture may be composed of one cell layer (single layer) or composed of two or more cell layers (laminate (multilayer), for example, two or three layers, Four layers, five layers, six layers, etc.). Further, the sheet-shaped cell culture may have a three-dimensional structure having a thickness exceeding the thickness of one cell without the cells showing a clear layer structure. For example, in the vertical cross section of the sheet-shaped cell culture, the cells may not be uniformly arranged in the horizontal direction, but may be non-uniformly arranged (for example, in a mosaic).
  • the implants of the present disclosure especially sheet cell cultures, preferably do not contain a scaffold. Scaffolds are sometimes used in the art to attach cells on and / or to their surfaces and maintain the physical integrity of sheet cell cultures, such as polyvinylidene difluoride ( Although PVDF) membranes and the like are known, the implants of the present disclosure can maintain their physical integrity without such a scaffold.
  • the sheet-shaped cell culture of the present disclosure preferably includes only a substance derived from the cells constituting the graft, and does not include any other substances.
  • the cell may be a cell derived from a different species or a cell derived from the same species.
  • heterologous cell means a cell derived from an organism of a different species from the recipient when a sheet-shaped cell culture is used for transplantation.
  • cells derived from monkeys and pigs correspond to xenogeneic cells.
  • Allogeneic cell means a cell derived from an organism of the same species as the recipient.
  • human cells correspond to cells derived from the same species.
  • Allogeneic cells include autologous cells (also called autologous cells or autologous cells), that is, cells derived from the recipient and allogeneic non-autologous cells (also called allogeneic cells). Autologous cells are preferred in the present disclosure because rejection does not occur even when transplanted. However, it is also possible to use xenogeneic cells or allogeneic non-autologous cells. When xenogeneic cells or allogeneic non-autologous cells are used, immunosuppressive treatment may be necessary to suppress rejection.
  • cells other than autologous cells that is, non-autologous cells of the same species as cells of xenogeneic origin may be collectively referred to as non-autologous cells.
  • the cells are autologous cells or allogeneic cells. In one aspect of the present disclosure, the cells are autologous cells (including autologous iPS cells). In another aspect of the present disclosure, the cells are allogeneic cells (including allogeneic iPS cells).
  • Cells constituting the graft of the present disclosure are cells that have been induced to differentiate from pluripotent stem cells, and are not particularly limited as long as they can form a graft such as a sheet-shaped cell culture.
  • Cells include, for example, adherent somatic cells (eg, cardiomyocytes, fibroblasts, epithelial cells, endothelial cells, hepatocytes, pancreatic cells, kidney cells, adrenal cells, periodontal ligament cells, gingival cells, periosteal cells, skin Cells, synovial cells, chondrocytes, etc.) and stem cells (eg, tissue stem cells such as myoblasts, cardiac stem cells, mesenchymal stem cells, etc.).
  • adherent somatic cells eg, cardiomyocytes, fibroblasts, epithelial cells, endothelial cells, hepatocytes, pancreatic cells, kidney cells, adrenal cells, periodontal ligament cells, gingival cells, periosteal cells, skin
  • the somatic cells may be stem cells, particularly those differentiated from iPS cells (iPS cell-derived adherent cells).
  • iPS cell-derived adherent cells include, for example, iPS cell-derived cardiomyocytes, fibroblasts, epithelial cells, endothelial cells, hepatocytes, pancreatic cells, kidney cells, adrenal cells, periodontal ligament cells Gingival cells, periosteal cells, skin cells, synovial cells, chondrocytes and the like.
  • the cells constituting the graft can be derived from any organism that can be treated with the graft. Such organisms include, without limitation, humans, non-human primates, dogs, cats, pigs, horses, goats, sheep, rodents (eg, mice, rats, hamsters, guinea pigs, etc.), rabbits, etc. Is included.
  • the number of types of cells constituting the graft is not particularly limited, and may be composed of only one type of cell, or may be a type using two or more types of cells.
  • the content ratio (purity) of the most abundant cells is, for example, 50% or more, preferably 60% or more, more preferably 70% or more at the end of the graft formation. More preferably, it can be 75% or more.
  • the culture substrate is not particularly limited as long as cells can form a cell culture thereon, and includes, for example, containers of various materials and / or shapes, a solid or semi-solid surface in the container, and the like.
  • the container is preferably made of a structure / material that does not allow the passage of a liquid such as a culture solution. Examples of such a material include, but are not limited to, polyethylene, polypropylene, Teflon (registered trademark), polyethylene terephthalate, polymethyl methacrylate, nylon 6,6, polyvinyl alcohol, cellulose, silicon, polystyrene, glass, polyacrylamide, and polydimethyl.
  • Acrylamide, metal for example, iron, stainless steel, aluminum, copper, brass
  • metal for example, iron, stainless steel, aluminum, copper, brass
  • the container preferably has at least one flat surface.
  • a culture container having a bottom surface formed of a culture substrate capable of forming a cell culture and a liquid impermeable side surface.
  • culture vessels include, but are not limited to, cell culture dishes, cell culture bottles, and the like.
  • the bottom of the container may be transparent or opaque. If the bottom surface of the container is transparent, observation and counting of cells can be performed from the back side of the container.
  • the container may have a solid or semi-solid surface inside. Examples of the solid surface include plates and containers made of various materials as described above, and examples of the semi-solid surface include a gel and a soft polymer matrix.
  • the culture substrate may be prepared using the above materials, or a commercially available substrate may be used.
  • Preferred culture substrates include, but are not limited to, for example, a substrate having an adhesive surface suitable for forming a sheet-shaped cell culture, and a substrate having a low adhesive surface suitable for forming a spheroid. And / or a substrate having a uniform well-like structure.
  • a hydrophilic compound such as a collagen gel or a hydrophilic polymer
  • collagen And extracellular matrices such as fibronectin, laminin, vitronectin, proteoglycan, and glycosaminoglycan
  • substrates coated on the surface with cell adhesion factors such as cadherin family, selectin family, and integrin family.
  • such substrates are commercially available (e.g., Corning (R) TC-Treated Culture Dish, Corning , etc.).
  • temperature-responsive gel obtained by crosslinking soft agar, poly (N-isopropylacrylamide) (PIPAAm) with polyethylene glycol (PEG), polyhydroxyethyl methacrylate (A substrate coated with a non-cell-adhesive compound such as a hydrogel such as poly (HEMA) or 2-methacryloyloxyethylphosphorhoscholine (MPC) polymer and / or a substrate having a uniform uneven structure on the surface.
  • a non-cell-adhesive compound such as a hydrogel such as poly (HEMA) or 2-methacryloyloxyethylphosphorhoscholine (MPC) polymer and / or a substrate having a uniform uneven structure on the surface.
  • HEMA poly (HEMA) or 2-methacryloyloxyethylphosphorhoscholine
  • MPC 2-methacryloyloxyethylphosphorhoscholine
  • the culture substrate may be entirely or partially transparent or opaque.
  • the culture substrate may be coated on its surface with a material whose properties change in response to a stimulus, for example, temperature or light.
  • materials include, but are not limited to, for example, (meth) acrylamide compounds, N-alkyl-substituted (meth) acrylamide derivatives (eg, N-ethylacrylamide, Nn-propylacrylamide, Nn-propylmethacrylamide, N-isopropylacrylamide, N-isopropylmethacrylamide, N-cyclopropylacrylamide, N-cyclopropylmethacrylamide, N-ethoxyethylacrylamide, N-ethoxyethylmethacrylamide, N-tetrahydrofurfurylacrylamide, N-tetrahydrofurfuryl methacryl Amide), N, N-dialkyl-substituted (meth) acrylamide derivatives (eg, N, N-dimethyl (meth) acrylamide, N, N-e
  • a predetermined stimulus By applying a predetermined stimulus to these materials, their physical properties, for example, hydrophilicity or hydrophobicity, can be changed, and the detachment of the cell culture adhered on the materials can be promoted.
  • Culture dishes coated with a temperature-responsive materials are commercially available (e.g., UpCell of CellSeed Inc. (R)), they can be used in the production method of the present disclosure.
  • the culture substrate may be in various shapes.
  • the area is not particularly limited, but may be, for example, about 1 cm 2 to about 200 cm 2 , about 2 cm 2 to about 100 cm 2 , about 3 cm 2 to about 50 cm 2 , and the like.
  • a circular culture dish having a diameter of 10 cm is used as a culture substrate.
  • the area is 56.7 cm 2 .
  • the culture surface may be flat or may have an uneven structure. In the case of having an uneven structure, it is preferable to have a uniform uneven structure.
  • inducible pluripotent stem cells or “iPS cells” are terms well known in the art, and by introducing multiple genes into somatic cells, three germ layers, that is, endoderm, mesoderm and A cell that has acquired the ability to differentiate into all lineage cells belonging to the ectoderm. That is, induced pluripotent stem cells or iPS cells are cells having pluripotency and self-renewal ability induced by introducing a gene.
  • the iPS cells are first cultured in suspension to form aggregates of any of the above three germ layers, and then the cells that form the aggregates are identified. Cells are induced to differentiate.
  • the iPS cells can be any mammalian cells, but are preferably iPS cells derived from human cells.
  • differentiation-inducing cells derived from iPS cells means any cells that have been subjected to differentiation-inducing treatment so as to differentiate from iPS cells into cells of a specific type.
  • Non-limiting examples of differentiation-inducing cells include muscular cells such as cardiomyocytes and skeletal myoblasts, neuronal cells such as neuronal cells, oligodendrocytes and dopamine-producing cells, retinal cells such as retinal pigment epithelial cells, and blood cells.
  • hematopoietic cells such as bone marrow cells
  • immune cells such as T cells, NK cells, NKT cells, dendritic cells, B cells, cells constituting organs such as hepatocytes, pancreatic ⁇ cells, kidney cells,
  • progenitor cells and somatic stem cells that differentiate into these cells are included.
  • progenitor cells and somatic stem cells include, for example, mesenchymal stem cells in cardiomyocytes, pluripotent heart progenitor cells, unipotent heart progenitor cells, neural stem cells in nervous system cells, hematopoietic cells and immune cells.
  • hematopoietic stem cells and lymphoid stem cells.
  • Induction of differentiation of iPS cells can be performed using any known technique. For example, differentiation induction from iPS cells to cardiomyocytes can be performed based on the method described in Miki et al., Cell Stem Cell 16, 699-711, June 4, 2015, and WO 2014/185358.
  • the differentiation-inducing cell may be a cell derived from an iPS cell into which any useful gene other than a gene for reprogramming has been introduced.
  • Non-limiting examples of such cells include, for example, iPS cells into which the gene for the chimeric antigen receptor described in Themeli M. et al. Nature Biotechnology, vol. 31, vol. 10, pp. 928-933, 2013 has been introduced. And T cells derived therefrom.
  • cells into which any useful gene has been introduced after differentiation induction from iPS cells are also included in the differentiation-inducing cells of the present invention.
  • One aspect of the present disclosure relates to a method for producing a high-quality sheet-shaped cell culture containing cardiomyocytes differentiated from iPS cells.
  • the method of the present disclosure includes the following steps (a) and (b): (A) a step of seeding a cell population containing cardiomyocytes differentiated from iPS cells on a culture substrate; and (b) a sheet culture of the seeded cell population with a sheeting medium containing a platelet lysate. Process.
  • cardiomyocytes means cells having characteristics of cardiomyocytes.
  • the characteristics of the cardiomyocyte include, but are not limited to, for example, the expression of a cardiomyocyte marker, the presence of an autonomous beat, and the like.
  • Non-limiting examples of cardiomyocyte markers include, for example, c-TNT (cardiac troponin T), CD172a (alias SIRPA or SHPS-1), KDR (alias CD309, FLK1 or VEGFR2), PDGFRA, EMILIN2, VCAM and the like.
  • the iPS cell-derived cardiomyocytes are c-TNT positive and / or CD172a positive.
  • the seeding on the culture substrate may be performed, for example, by injecting a cell suspension in which cells are suspended in a sheeting medium into a culture container provided with the culture substrate.
  • a device suitable for the operation of injecting the cell suspension such as a dropper or pipette, can be used.
  • the seeding density of the cells is determined at a density capable of forming a sheet-shaped cell culture, and the density may vary depending on the desired cells. However, those skilled in the art can select an appropriate density from techniques known in the art. can do.
  • a sheet-shaped cell culture containing cardiomyocytes it may be, for example, 2.0 ⁇ 10 5 cells / cm 2 or more, but may be seeded at a higher density.
  • Examples of higher densities include, for example, densities that reach confluence, i.e., densities at which cells are expected to cover the entire adhesive surface of the culture vessel upon seeding, e.g., upon seeding, cells contact each other , A density at which contact inhibition occurs, or a density at which cell growth is substantially stopped by contact inhibition.
  • the upper limit of the seeding density is not particularly limited. However, if the seeding density is excessively high, the number of dead cells increases, resulting in inefficiency.
  • the seeding density is, for example, from about 1.0 ⁇ 10 6 / cm 2 to about 1.0 ⁇ 10 7 / cm 2 , from about 1.0 ⁇ 10 6 / cm 2 to about 5 0.0 ⁇ 10 6 / cm 2 , about 1.0 ⁇ 10 6 / cm 2 to about 3.0 ⁇ 10 6 / cm 2 , about 1.5 ⁇ 10 6 / cm 2 to about 1.0 ⁇ 10 7 pieces / cm 2 , about 1.5 ⁇ 10 6 pieces / cm 2 to about 5.0 ⁇ 10 6 pieces / cm 2 , about 1.5 ⁇ 10 6 pieces / cm 2 to about 3.0 ⁇ 10 6 pieces / cm 2 , about 2.0 ⁇ 10 6 pieces / cm 2 to about 1.0 ⁇ 10 7 pieces / cm 2 , about 2.0 ⁇ 10 6 pieces / cm 2 to about 5.0 ⁇ 10 6 pieces / Cm 2 , about 2.0 ⁇ 10 6 / cm 2 to about 3.0 ⁇ 10 6 / cm 2 , and the like.
  • the seeding density is, for
  • the culture substrate to be seeded is as described in detail above, but in a preferred embodiment, is a culture substrate having a surface coated with a cell adhesive component such as an extracellular matrix or a cell adhesion factor.
  • Cell adhesion components include, but are not limited to, for example, extracellular matrices such as collagen, fibronectin, laminin, vitronectin, proteoglycans, glycosaminoglycans, cadherin family, selectin family, cell adhesion factors such as integrin family And modifications thereof, for example, laminin 511 (a modification of laminin), VTN-N (a modification of vitronectin), and RetroNectin (R) (a modification of fibronectin).
  • the seeded cell population may contain other cells as long as it contains cardiomyocytes differentiated from iPS cells.
  • a cell population obtained by inducing differentiation from iPS cells into cardiomyocytes may include, for example, fibroblasts and vascular endothelial cells in addition to cardiomyocytes.
  • a cell population obtained by inducing differentiation from iPS cells using the method described in Miki et al., Cell Stem Cell 16, 699-711, June 4, 2015 or WO 2014/185358, or the like is used as it is. It may be used, or may be used after carrying out cryopreservation or pre-culture.
  • the cell population to be seeded is induced to differentiate from iPS cells, and then inoculated on a culture substrate (preferably on a planar culture substrate) to perform adherent culture. is there. Cryopreservation and thawing may be performed before or after such adherent culture. By performing the adherent culture, it is possible to achieve the formation of a high-quality graft at a high probability in the subsequent formation of the graft.
  • the formed graft may include vascular endothelial cells, cell walls, fibroblasts in addition to cardiomyocytes.
  • the percentage of cells contained in the graft of the present disclosure may be, for example, about 30-70% of cardiomyocytes, 0.1% to about 20% of vascular endothelial cells, and about 1% to about 40% of mural cells. Good.
  • the culture conditions and the like may be in accordance with the conditions for performing ordinary adhesion culture.
  • the culture may be performed at 37 ° C. under 5% CO 2 using a commercially available culture vessel for adhesion culture.
  • the seeding density of the cells may be any density as long as the density does not prevent adhesion between cells and / or formation of adhesion between the cells and the culture substrate, for example, a subconfluent density, The density may be confluent or higher.
  • the cultivation time may be a time period such that adhesion between cells and / or adhesion between cells and a culture substrate is formed, and specifically, for example, 2 to 168 hours, 2 to 144 hours, 2 to 120 hours It may be about 2 to 96 hours, 2 to 72 hours, 2 to 48 hours, 2 to 24 hours, 2 to 12 hours, 2 to 6 hours, 2 to 4 hours.
  • the seeded cells are cultured on a sheet.
  • a culture for forming the seeded cells as a graft is referred to as a “graft formation culture”, and the graft is a sheet-shaped cell culture, and the culture for forming the seeded cells into a sheet is performed.
  • sheet culture it is referred to as “sheet culture”. Sheeting of the seeded cells can be performed by any known method and conditions. Non-limiting examples of such techniques are described in, for example, JP-A-2010-081829, JP-A-2010-226991, JP-A-2011-110368, JP-A-2011-172925, WO 2014/185517, and the like.
  • the step of forming the seeded cells into a sheet can be achieved, for example, by culturing the cells under conditions that form intercellular adhesion.
  • Such conditions may be any as long as they can form cell-cell adhesion, but usually, cell-cell adhesion can be formed under the same conditions as general cell culture conditions.
  • Such conditions include, for example, culture at about 37 ° C., 5% CO 2 .
  • the cultivation can be performed under normal pressure (atmospheric pressure, non-pressurized).
  • the culture can be performed in a container of any size and shape.
  • the size and shape of the sheet-shaped cell culture can be adjusted by adjusting the size and shape of the cell attachment surface of the culture container, or by installing a mold of the desired size and shape on the cell attachment surface of the culture container, It can be adjusted arbitrarily by culturing the cells therein.
  • the time of sheeting culture may vary depending on the type of seeded cells and cell density.
  • the sheets may be seeded at a density of, for example, about 2.1 ⁇ 10 5 cells / cm 2 and cultured for 4 days or more to form sheets.
  • the seeding density reaches a confluent density, that is, when the seeding is performed at a higher density, the period of sheet culture can be shortened, and the culture time may be 2 to 4 days, more preferably 2 to 3 days. .
  • a medium used for graft formation for example, sheeting
  • a graft forming medium sometimes referred to as a sheeting medium particularly when the graft formation is a sheeting
  • a medium capable of forming a cell graft is used.
  • physiological saline various physiological buffers (for example, PBS, HBSS, etc.), or those based on various cell culture base media.
  • basal medium examples include, but are not limited to, DMEM, MEM, F12, DME, RPMI1640, MCDB (MCDB102, 104, 107, 120, 131, 153, 199, etc.), L15, SkBM, RITC80-7, DMEM / F12 and the like.
  • MCDB MCDB102, 104, 107, 120, 131, 153, 199, etc.
  • L15 SkBM, RITC80-7
  • DMEM / F12 examples include, but are not limited to, DMEM, MEM, F12, DME, RPMI1640, MCDB (MCDB102, 104, 107, 120, 131, 153, 199, etc.), L15, SkBM, RITC80-7, DMEM / F12 and the like.
  • the basal medium may be used with its standard composition (for example, as it is commercially available), or its composition may be appropriately changed depending on the cell type and cell conditions. Therefore, the
  • the graft forming medium may contain additives such as normal serum (eg, bovine serum such as fetal bovine serum, horse serum, human serum, etc.) and various growth factors (eg, FGF, EGF, VEGF, HGF, etc.).
  • normal serum eg, bovine serum such as fetal bovine serum, horse serum, human serum, etc.
  • various growth factors eg, FGF, EGF, VEGF, HGF, etc.
  • the transplant contains no serum different from the species from which the cells contained in the transplant are derived, such as bovine serum and horse serum.
  • the present disclosure is characterized in that a graft forming culture is performed using a graft forming medium containing a platelet lysate instead of or in addition to serum or a growth factor.
  • the graft forming medium comprises platelet lysate but does not contain serum.
  • platelet lysate (PL) refers to a composition rich in growth factors and the like, obtained by repeatedly freezing and thawing platelets.
  • platelet lysates have been known to promote the growth of mesenchymal stem cells.
  • the present inventors have added a platelet lysate to a sheeting medium, thereby achieving a strong autonomic pulsation in a shorter culture period than before. Was observed for the first time.
  • Platelet lysates are commercially available as media additives for cell culture and are known in the art.
  • the platelet lysate can be prepared, for example, by the method described in JP-T-2014-533715, Bieback et al., STEM CELLS, 2009; 27: 2331-2341.
  • it can be prepared by, for example, dissolving a platelet population and removing contaminants such as platelet particles and a membrane therefrom to obtain a supernatant. Lysis of platelets can be achieved through steps such as chemical means (eg, using CaCl 2 ), osmotic means (eg, using distilled water), freeze-thaw means, mechanical disruption means, and the like. Removal of contaminants can be achieved by a method such as centrifugation or filtration.
  • the concentration of the platelet lysate contained in the graft forming medium may be a level generally used in the art, such as 1%, 2.5%, 5%, 10%, 15%, 20%, and the like. May be.
  • the platelet lysate is contained in the graft forming medium in an amount of about 1% to 20%, more preferably about 2% to 10%, and still more preferably about 2.5% to 10%.
  • the graft forming medium may be appropriately replaced during the graft forming culture. Further, the composition of the medium may be changed in accordance with the progress of the graft formation.
  • the present inventors use a sheeting medium to which a Rho kinase (ROCK) inhibitor is added as a medium on the first day of sheeting culture in the production of a sheet-shaped cell culture containing cardiomyocytes derived from iPS cells. As a result, it was newly found that a sheet-shaped cell culture was effectively formed.
  • the sheeting medium used for day 1 sheeting culture comprises a Rho kinase inhibitor.
  • the sheeting medium after the second day may or may not contain a Rho kinase inhibitor, but preferably does not contain a Rho kinase inhibitor.
  • the sheeting medium may further contain a cell adhesive component.
  • the cell adhesive component is as described in detail above.
  • the culture substrate may or may not be coated with the cell adhesive component.
  • the cell adhesive component contained in the sheeting medium may be the same as or different from the cell adhesive component coating the culture substrate. However, they are preferably the same cell adhesive component.
  • the concentration of the cell-adhesive component contained in the graft-forming medium may vary depending on the type of the cell-adhesive component contained, the state of the cells forming the graft, and the like. For example, when cells having low viability, that is, cells having weak activity, are used, the content of the cell adhesive component is preferably small.
  • the concentration of the cell adhesive component contained in the sheeting medium is about 0.1% and about 0.1% based on the concentration (100%) used when the same cell adhesive component is used as a coating agent for the culture substrate. It may be 5%, about 1%, about 5%, about 10%, about 20%, about 25%, about 50%, about 75%, about 100%, etc.
  • the concentration range of the cell adhesive component contained in the sheeting medium is about 0% based on the concentration (100%) used when the same cell adhesive component is used as a coating agent for the culture substrate. 0.1% to about 100%, about 0.1% to about 100%, about 0.1% to about 50%, about 0.1% to about 25%, about 0.1% to about 20%, about 0% 0.1% to about 10%, about 1% to about 100%, about 1% to about 100%, about 0.5% to about 100%, about 0.5% to about 100%, about 0.5% to About 50%, about 0.5% to about 25%, about 0.5% to about 20%, about 0.5% to about 10%, about 1% to about 50%, about 1% to about 25%, About 1% to about 20%, about 1% to about 10%, about 0.5% to about 100%, about 5% to about 100%, about 5% to about 50%, about 5% to about 25%, About 5% to about 20%, May the like 5% to about 10%.
  • Another aspect of the present disclosure is a method of hastening the onset of pulsation of cardiomyocytes in a graft containing cardiomyocytes differentiated from iPS cells, comprising:
  • the present invention relates to the above method, comprising culturing the explant in a formation medium.
  • the present inventors compared platelet formation with a serum-free medium by adding a platelet lysate to a graft formation medium when forming a graft containing cardiomyocytes differentiated from iPS cells. It was found that the onset of pulsation of cardiomyocytes was accelerated. This makes it possible to easily form a high-quality implant in a xeno-free environment.
  • Another aspect of the present disclosure relates to a method of treating a disease in a subject, comprising applying an effective amount of an implant produced by the method of the present disclosure to the subject in need thereof.
  • the disease to be treated is as described above.
  • treatment is intended to include all types of medically acceptable prophylactic and / or therapeutic interventions, such as for the cure, temporary remission or prevention of disease.
  • treatment includes medically acceptable treatments for a variety of purposes, including slowing or stopping the progression of a disease associated with tissue abnormalities, regressing or eliminating lesions, preventing the onset of the disease or preventing its recurrence, and the like. Involve interventions.
  • a component that enhances the survival, engraftment, and / or function of a graft, and other active components that are useful for treating a target disease are used in combination with the graft or the like of the present disclosure. be able to.
  • the treatment method of the present disclosure may further include a step of manufacturing the implant of the present disclosure according to the manufacturing method of the present disclosure.
  • the method of treatment of the present disclosure may comprise, prior to the step of producing the graft, cells (eg, somatic cells for inducing iPS cells, such as skin cells, blood cells, etc.) or cells for producing the graft from the subject.
  • the method may further include a step of collecting a tissue (for example, a tissue containing a somatic cell for inducing iPS cells, such as skin tissue or blood) serving as a source of the protein.
  • a tissue for example, a tissue containing a somatic cell for inducing iPS cells, such as skin tissue or blood
  • the subject from whom the cells or tissue from which the cells are to be sourced is harvested is the same individual as the subject to whom a cell culture, composition, or explant is administered.
  • the subject from whom the cells or tissue from which the cells are to be sourced is harvested is a homologous distinct body from the subject to be administered, such as a cell culture, composition, or implant.
  • the subject from which the cells or the tissue from which the cells are sourced is harvested is an individual that is heterogeneous to the subject receiving the administration, such as a graft.
  • an effective amount is, for example, an amount capable of suppressing the onset or recurrence of a disease, reducing symptoms, or delaying or stopping the progress (eg, size, weight, number, etc. of a graft), Preferably, it is an amount that prevents the onset and recurrence of the disease or cures the disease. Also preferred is an amount that does not cause adverse effects beyond the benefit of administration. Such an amount can be appropriately determined, for example, by a test in a laboratory animal such as a mouse, a rat, a dog or a pig, or a disease model animal, and such a test method is well known to those skilled in the art.
  • the size of a tissue lesion to be treated can be an important index for determining an effective amount.
  • the administration method examples include intravenous administration, intramuscular administration, intraosseous administration, intrathecal administration, and direct application to tissues.
  • the frequency of administration is typically once per treatment, but multiple administrations are possible if the desired effect is not obtained.
  • the cell culture, the composition, the sheet-shaped cell culture, or the like of the present invention may be fixed to a target tissue by a locking means such as a suture or staple.
  • human iPS cells for clinical use established at Kyoto University iPS Cell Research Institute were used as pluripotent stem cells.
  • Human iPS cells were maintained by a feeder-free method with reference to M. Nakagawa et al., Scientific Reports, 4: 3594 (2014).
  • human iPS cells were induced to differentiate into cardiomyocytes by referring to Miki et al., Cell Stem Cell 16, 16, 699-711, June 4, 2015 and WO 2014/185358 and WO 2017/038562 to induce embryoid bodies.
  • the cells were cultured in a culture solution containing the agent (Dorsomorphin) and a TGF ⁇ inhibitor (SB431542), and then cultured in a culture solution containing VEGF and bFGF to obtain iPS cell-derived human cardiomyocytes.
  • the percentage of cardiomyocytes in the resulting cell population was between 50% and 90%.
  • Example 1 Comparison between FBS-containing medium and PL-containing medium Using the cell population containing cardiomyocytes differentiated from human iPS cells obtained above, sheet-forming culture conditions were examined.
  • a sheet medium was prepared by adding 20% FBS or 5% human platelet lysate to DMEM / F12 medium.
  • laminin iMatrix-5111 was further added as a cell adhesive component at 0.1 ⁇ g / mL, 0.25 ⁇ g / mL or 0.5 ⁇ g / mL, respectively.
  • the Rho kinase inhibitor Y27632 was added to the sheeting medium.
  • Cell population comprising cardiomyocytes, temperature responsive culture dishes at a density of 1.5 ⁇ 10 6 cells / cm 2 (UpCell (R) , Cellseed) seeded, 37 ° C., 3 days of culture in the 5% CO 2 environment did.
  • the temperature-responsive culture dish used was the same as the culture solution (excluding Y27632), and was incubated at 37 ° C. overnight and precoated. After the culture, the sheet-shaped cell culture containing cardiomyocytes was detached from the culture dish.
  • Example 2 Similar to Comparative Example 1 between serum-free medium and PL-containing medium , DMEM / F12 medium supplemented with 0.1 ⁇ g / mL laminin (iMatrix-511), and further supplemented with 5% platelet lysate Were used as sheeting media to perform sheeting culture. The results are shown in FIG. Comparing the sheeting success rate on the second day of sheeting culture, the PL group was 100% successful, whereas the serum-free medium was 50%.
  • the state of the pulsation was photographed using the cell motion imaging system SI8000 (Sony), and the pulsation acceleration (acceleration) and pulsation deformation distance ( contration deformation distance) was calculated and compared between the case where the FBS-containing medium of Example 1 was used and the case where the PL-containing medium of Example 2 was used.
  • the results are shown in FIG. It was confirmed that the group in which the sheet-shaped cell culture was formed using the PL-containing medium had a stronger pulsation than the group using the FBS-containing medium.
  • a high-quality sheet-shaped cell culture can be obtained when a sheet-shaped cell culture is formed using cells or the like that have been induced to differentiate from pluripotent stem cells.
  • a high-quality sheet cell culture can be easily formed.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Cell Biology (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Zoology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Cardiology (AREA)
  • Epidemiology (AREA)
  • Developmental Biology & Embryology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Transplantation (AREA)
  • Vascular Medicine (AREA)
  • Immunology (AREA)
  • Virology (AREA)
  • Oral & Maxillofacial Surgery (AREA)
  • Hospice & Palliative Care (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Botany (AREA)
  • Dermatology (AREA)
  • Surgery (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Materials For Medical Uses (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The purpose of the present invention is to provide: a method for producing a high-quality graft, such as a sheet-shaped cell culture, that includes a variety of iPS cell-derived cells, such as cardiomyocytes, while possessing the functions of the cells to a high degree; a graft produced by using the method; and a method for treating a disease by using the graft. The problem is solved by a method for producing a graft, the method comprising: (a) a step for inoculating a cell population that includes cells differentiation-induced from an iPS cell onto a culture substrate, and (b) a step for performing a graft-forming cultivation of the inoculated cell population on a graft formation medium that includes a platelet lysate.

Description

心筋細胞のシート化方法Cardiomyocyte sheeting method
 本開示は、平成30年度、国立研究開発法人日本医療研究開発機構 再生医療実現拠点ネットワークプログラム 疾患・組織別実用化研究拠点(拠点A)「iPS細胞を用いた心筋再生治療創成拠点」に係る委託研究開発、産業技術力強化法第19条の適用を受ける特許出願であって、人工多能性幹細胞由来の分化誘導細胞、特に心筋細胞を含む、シート状細胞培養物などの移植片を製造する方法、当該方法を用いて製造された移植片、当該移植片を用いた疾患の処置方法などに関する。 This disclosure was concluded in FY2018 by the Japan Agency for Medical Research and Development, Japan Network Program for the Realization of Regenerative Medicine, Research Center for Practical Use by Disease / Organization (Center A), "Creation Center for Myocardial Regenerative Therapy Using iPS Cells" A patent application subject to Article 19 of the Research and Development, Industrial Technology Enhancement Act, which produces transplants, such as sheet-shaped cell cultures, including differentiation-inducing cells derived from induced pluripotent stem cells, particularly cardiomyocytes. The present invention relates to a method, a graft manufactured using the method, a method for treating a disease using the graft, and the like.
 成体の心筋細胞は自己複製能に乏しく、心筋組織が損傷を受けた場合、その修復は極めて困難である。近年、損傷した心筋組織の修復のために、細胞工学的手法により作製した心筋細胞を含む移植片を患部に移植する試みが行われている(特許文献1、非特許文献1)。かかる移植片の作製に用いる心筋細胞の給源として最近注目されているのが、胚性幹細胞(ES細胞)や人工多能性幹細胞(iPS細胞)などの多能性幹細胞から誘導した心筋細胞であり、このような多能性幹細胞由来の心筋細胞を含むシート状細胞培養物の作製や動物での治療実験が試みられている(非特許文献2~3)。しかしながら、多能性幹細胞由来の心筋細胞を含むシート状細胞培養物の開発は始まったばかりであり、その機能的特性や、それに影響する因子などについては依然不明な部分が多い。 Adult cardiomyocytes have poor self-renewal ability, and repair of damaged myocardial tissue is extremely difficult. In recent years, in order to repair damaged myocardial tissue, attempts have been made to transplant a graft containing cardiomyocytes prepared by a cell engineering technique into an affected part (Patent Document 1, Non-Patent Document 1). As a source of cardiomyocytes used for preparing such grafts, cardiomyocytes derived from pluripotent stem cells such as embryonic stem cells (ES cells) and induced pluripotent stem cells (iPS cells) have recently been receiving attention. Production of sheet-shaped cell cultures containing such pluripotent stem cell-derived cardiomyocytes and treatment experiments on animals have been attempted (Non-patent Documents 2 and 3). However, the development of sheet-like cell cultures containing cardiomyocytes derived from pluripotent stem cells has only just begun, and there are still many unknowns regarding their functional properties and factors affecting them.
 再生医療に用いる臨床用のシート状細胞培養物を製造する場合、なるべくヒト以外の異種由来成分を含まない状態、いわゆるゼノフリーな状態で製造されることが望ましい。しかしながら、ゼノフリーな状態でシート状細胞培養物を形成することはコストや手間がかかる場合が多く、また用いる細胞によっては該細胞の所望の性質を保ったままシート状細胞培養物を形成することが困難である場合もある。 (4) When producing a sheet-shaped cell culture for clinical use used in regenerative medicine, it is desirable that the cell-shaped cell culture be produced in a state containing as little as possible foreign components other than humans, that is, in a so-called xeno-free state. However, forming a sheet-shaped cell culture in a xeno-free state is often costly and time-consuming, and depending on the cells used, it may be necessary to form a sheet-shaped cell culture while maintaining the desired properties of the cells. It can be difficult.
特表2007-528755号公報JP-T 2007-528755
 本開示は、iPS細胞由来の分化誘導細胞、特に心筋細胞、を該細胞の機能を高度に有したまま含み、高品質なシート状細胞培養物などの移植片を製造する方法、当該方法を用いて製造された移植片、当該移植片を用いた疾患の処置方法などを提供することを目的とする。 The present disclosure relates to a method for producing a transplant such as a high-quality sheet-shaped cell culture, which includes iPS cell-derived differentiation-inducing cells, particularly cardiomyocytes, while maintaining the function of the cells at a high level, and using the method. It is an object of the present invention to provide a graft manufactured by the method, a method for treating a disease using the graft, and the like.
 移植の用に供する心筋細胞を含む移植片を調製するにあたっては、ゼノフリー環境下で調製される必要がある。また、ウシ胎児血清(FBS)を用いたり無血清培地を用いたりすると、心筋細胞が十分に拍動しないなどの問題が見出されていた。 に あ た っ て When preparing a graft containing cardiomyocytes to be used for transplantation, it must be prepared in a xeno-free environment. In addition, when using fetal bovine serum (FBS) or a serum-free medium, a problem such as insufficient pulsation of cardiomyocytes has been found.
 本発明者らは、ゼノフリー環境下で調製しても十分な機能を有するiPS細胞由来の心筋細胞を用いた生体移植用のシート状細胞培養物の調製を試みる中で、血清の代わりに血小板溶解物(Platelet lysate)を用いると、血清を用いた場合よりも短時間で強い拍動を観察できるという新たな知見を見出した。かかる知見に基づいてさらに研究を進め、iPS細胞から分化誘導された種々の体細胞を含有する移植片の調製において、臨床応用に耐え得る高品質な移植片を製造することが可能であることを見出し、本発明を完成させるに至った。 The present inventors have attempted to prepare a sheet-shaped cell culture for living body transplantation using iPS cell-derived cardiomyocytes having a sufficient function even if prepared in a xenofree environment, Using a platelet (lysate), a new finding was found that a stronger pulsation could be observed in a shorter time than when serum was used. Based on such findings, further research has been carried out to demonstrate that it is possible to produce high-quality transplants that can withstand clinical applications in the preparation of transplants containing various somatic cells differentiated from iPS cells. As a result, the present invention has been completed.
 すなわち、本発明に下記に掲げるものに関する:
[1]多能性幹細胞から分化誘導された細胞を含む移植片を製造する方法であって;
(a)前記細胞を含む細胞集団を、培養基材上に播種する工程、および
(b)播種した細胞集団を、血小板溶解物を含む移植片形成媒体で移植片形成培養する工程、
を含む、前記方法。
[2]多能性幹細胞が、iPS細胞である、[1]の方法。
[3]移植片が、シート状細胞培養物である、[1]または[2]の方法。
[4]移植片形成媒体が、移植片に含まれる細胞種と異種の血清を含まない、[1]~[3]の方法。
[5]移植片形成媒体が、さらに細胞接着性成分を含む、[1]~[4]の方法。
[6]培養基材が、細胞接着性成分および/または血小板溶解物でコーティングされている、[1]~[5]の方法。
[7]細胞が、心筋細胞である、[1]~[6]の方法。
[8]iPS細胞から分化誘導した心筋細胞を含む移植片において該心筋細胞の拍動開始を早める方法であって、前記心筋細胞を含む細胞集団を、血小板溶解物を含む移植片形成媒体で移植片形成培養することを含む、前記方法。
That is, the present invention relates to the following:
[1] A method for producing a transplant containing cells induced to differentiate from pluripotent stem cells;
(A) a step of inoculating a cell population containing the cells on a culture substrate, and (b) a step of explanting and culturing the inoculated cell population with an explant forming medium containing a platelet lysate;
The above method, comprising:
[2] The method of [1], wherein the pluripotent stem cells are iPS cells.
[3] The method of [1] or [2], wherein the transplant is a sheet-shaped cell culture.
[4] The method of [1] to [3], wherein the graft-forming medium does not contain serum different from the cell type contained in the graft.
[5] The method of [1] to [4], wherein the graft forming medium further contains a cell adhesive component.
[6] The method of [1] to [5], wherein the culture substrate is coated with a cell adhesive component and / or a platelet lysate.
[7] The method of [1] to [6], wherein the cells are cardiomyocytes.
[8] A method for accelerating the onset of pulsation of cardiomyocytes in a graft containing cardiomyocytes differentiated from iPS cells, wherein the cell population containing cardiomyocytes is transplanted with a graft-forming medium containing platelet lysate The above-described method, comprising culturing a piece.
 本発明によれば、iPS細胞から分化誘導した分化誘導細胞、特に心筋細胞を含む細胞集団から、従来よりも高品質なシート状細胞培養物などの移植片を高効率に製造することができる。特にiPS細胞から分化誘導した心筋細胞など、再生医療に用いるシート状細胞培養物などの移植片の調製において、所望の性質を高いレベルで保持した移植片を調製可能であり、とくにヒトへの生体移植用に非常に好適な移植片を提供することが可能となる。 According to the present invention, a transplant, such as a sheet-shaped cell culture, having a higher quality than before can be produced with high efficiency from a cell population including differentiation-inducing cells, particularly cardiomyocytes, that have been induced to differentiate from iPS cells. In particular, in the preparation of grafts such as sheet-shaped cell cultures used for regenerative medicine, such as cardiomyocytes differentiated from iPS cells, it is possible to prepare grafts retaining desired properties at a high level. It is possible to provide a graft which is very suitable for transplantation.
図1は、例2における無血清培地とPL含有培地とをそれぞれ用いてシート化培養を行った場合の結果の比較を表す表である。PL含有培地を用いた場合は、2日間の培養で全てのロットでシートが得られたが、無血清培地を用いた場合は、半分のロットで破損などによりシートができなかった。FIG. 1 is a table showing a comparison of the results when sheet culture was performed using the serum-free medium and the PL-containing medium in Example 2 respectively. When the PL-containing medium was used, sheets were obtained in all lots after two days of culture, but when the serum-free medium was used, sheets could not be formed due to breakage in half of the lots. 図2は、FBS含有培地を用いた場合と、PL含有培地を用いた場合とで作成されるシート状心筋細胞培養物の拍動の様子を比較したグラフである。PL含有培地で作成されたものは、FBS含有培地で作成されたものと比較して、早期に強い拍動が観察された。FIG. 2 is a graph comparing the state of pulsation of a sheet-shaped cardiomyocyte culture prepared using an FBS-containing medium and using a PL-containing medium. A stronger pulsation was observed earlier in the medium prepared with the PL-containing medium than in the medium prepared with the FBS-containing medium.
 以下、本発明を詳細に説明する。
 本明細書において別様に定義されない限り、本明細書で用いる全ての技術用語および科学用語は、当業者が通常理解しているものと同じ意味を有する。本明細書中で参照する全ての特許、出願および他の出版物や情報は、その全体を参照により本明細書に援用する。また本明細書において参照された出版物と本明細書の記載に矛盾が生じた場合は、本明細書の記載が優先されるものとする。
Hereinafter, the present invention will be described in detail.
Unless defined otherwise herein, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. All patents, applications and other publications and information referred to herein are hereby incorporated by reference in their entirety. In the case of inconsistencies between the publication referred to in this specification and the description of this specification, the description of this specification shall control.
 本開示は、多能性幹細胞から分化誘導された細胞を含む移植片を製造する方法であって;
(a)前記細胞を含む細胞集団を、培養基材上に播種する工程、および
(b)播種した細胞集団を、血小板溶解物を含む培地で移植片形成培養する工程、
を含む、前記方法に関する。
The present disclosure provides a method for producing an implant comprising cells induced to differentiate from pluripotent stem cells;
(A) inoculating a cell population containing the cells on a culture substrate, and (b) explanting and culturing the inoculated cell population in a medium containing platelet lysate;
The method relates to:
 本開示において、「移植片」とは、生体内へ移植するための構造物を意味し、特に細胞を構成成分として含む移植用構造物を意味する。移植片において細胞同士は接着して全体としてある形状を形成している状態を少なくとも一つ含み、一つ一つの細胞が全てバラバラに遊離して存在している、いわゆる懸濁状態は、本開示の「移植片」には含まれない。好ましい一態様においては、移植片は、細胞および細胞由来の物質以外の構造物(例えばスキャフォールドなど)を含まない移植用構造物である。本開示における移植片としては、これに限定するものではないが、例えばシート状細胞培養物、スフェロイド、細胞凝集塊、細胞懸濁物、フィブリンゲルを含む細胞懸濁物、ナノファイバーを用いた細胞培養物などが挙げられ、好ましくはシート状細胞培養物またはスフェロイド、より好ましくはシート状細胞培養物である。 に お い て In the present disclosure, the term “graft” refers to a structure for transplantation into a living body, and particularly refers to a structure for transplantation containing cells as a component. The so-called suspension state, in which at least one state in which cells are adhered to each other in a transplant to form a certain shape as a whole, and each and every cell is present separately, is referred to as the present disclosure. Are not included in the “grafts” In a preferred embodiment, the implant is an implantable structure that does not include structures other than cells and cell-derived substances (eg, a scaffold). Examples of the graft in the present disclosure include, but are not limited to, a sheet-shaped cell culture, a spheroid, a cell aggregate, a cell suspension, a cell suspension containing fibrin gel, and a cell using a nanofiber. Cultures and the like are preferable, and a sheet cell culture or a spheroid is preferable, and a sheet cell culture is more preferable.
 本開示において、「シート状細胞培養物」は、細胞が互いに連結してシート状になったものをいう。本開示において、「スフェロイド」は細胞が互いに連結して略球状になったものをいう。細胞同士は、直接(接着分子などの細胞要素を介するものを含む)および/または介在物質を介して、互いに連結していてもよい。介在物質としては、細胞同士を少なくとも物理的(機械的)に連結し得る物質であれば特に限定されないが、例えば、細胞外マトリックスなどが挙げられる。介在物質は、好ましくは細胞由来のもの、特に、シート状細胞培養物やスフェロイドを構成する細胞に由来するものである。細胞は少なくとも物理的(機械的)に連結されるが、さらに機能的、例えば、化学的、電気的に連結されてもよい。シート状細胞培養物は、1の細胞層から構成されるもの(単層)であっても、2以上の細胞層から構成されるもの(積層体(多層)、例えば、2層、3層、4層、5層、6層など)であってもよい。また、シート状細胞培養物は、細胞が明確な層構造を示すことなく、細胞1個分の厚みを超える厚みを有する3次元構造を有してもよい。例えば、シート状細胞培養物の垂直断面において、細胞が水平方向に均一に整列することなく、不均一に(例えば、モザイク状に)配置された状態で存在していてもよい。  に お い て In the present disclosure, the “sheet-shaped cell culture” refers to a cell in which cells are connected to each other to form a sheet. In the present disclosure, “spheroid” refers to a cell in which cells are connected to each other to form a substantially spherical shape. The cells may be connected to each other directly (including via a cellular element such as an adhesion molecule) and / or via an intermediary substance. The intervening substance is not particularly limited as long as it is a substance capable of at least physically (mechanically) connecting cells, and examples thereof include an extracellular matrix. The intervening substance is preferably derived from cells, particularly from cells constituting a sheet-shaped cell culture or spheroid. Cells are at least physically (mechanically) linked, but may be further functionally linked, eg, chemically or electrically. The sheet-shaped cell culture may be composed of one cell layer (single layer) or composed of two or more cell layers (laminate (multilayer), for example, two or three layers, Four layers, five layers, six layers, etc.). Further, the sheet-shaped cell culture may have a three-dimensional structure having a thickness exceeding the thickness of one cell without the cells showing a clear layer structure. For example, in the vertical cross section of the sheet-shaped cell culture, the cells may not be uniformly arranged in the horizontal direction, but may be non-uniformly arranged (for example, in a mosaic).
 本開示の移植片、特にシート状細胞培養物は、好ましくはスキャフォールド(支持体)を含まない。スキャフォールドは、その表面上および/またはその内部に細胞を付着させ、シート状細胞培養物の物理的一体性を維持するために当該技術分野において用いられることがあり、例えば、ポリビニリデンジフルオリド(PVDF)製の膜等が知られているが、本開示の移植片は、かかるスキャフォールドがなくともその物理的一体性を維持することができる。また、本開示のシート状細胞培養物は、好ましくは、移植片を構成する細胞由来の物質のみからなり、それら以外の物質を含まない。 移植 The implants of the present disclosure, especially sheet cell cultures, preferably do not contain a scaffold. Scaffolds are sometimes used in the art to attach cells on and / or to their surfaces and maintain the physical integrity of sheet cell cultures, such as polyvinylidene difluoride ( Although PVDF) membranes and the like are known, the implants of the present disclosure can maintain their physical integrity without such a scaffold. In addition, the sheet-shaped cell culture of the present disclosure preferably includes only a substance derived from the cells constituting the graft, and does not include any other substances.
 細胞は異種由来細胞であっても同種由来細胞であってもよい。ここで「異種由来細胞」は、シート状細胞培養物が移植に用いられる場合、そのレシピエントとは異なる種の生物に由来する細胞を意味する。例えば、レシピエントがヒトである場合、サルやブタに由来する細胞などが異種由来細胞に該当する。また、「同種由来細胞」は、レシピエントと同一の種の生物に由来する細胞を意味する。例えば、レシピエントがヒトである場合、ヒト細胞が同種由来細胞に該当する。同種由来細胞は、自己由来細胞(自己細胞または自家細胞ともいう)、すなわち、レシピエントに由来する細胞と、同種非自己由来細胞(他家細胞ともいう)を含む。自己由来細胞は、移植しても拒絶反応が生じないため、本開示においては好ましい。しかしながら、異種由来細胞や同種非自己由来細胞を利用することも可能である。異種由来細胞や同種非自己由来細胞を利用する場合は、拒絶反応を抑制するため、免疫抑制処置が必要となることがある。なお、本明細書中で、自己由来細胞以外の細胞、すなわち、異種由来細胞と同種非自己由来細胞を非自己由来細胞と総称することもある。本開示の一態様において、細胞は自家細胞または他家細胞である。本開示の一態様において、細胞は自家細胞(自家iPS細胞を含む)である。本開示の別の態様において、細胞は他家細胞(他家iPS細胞を含む)である。 The cell may be a cell derived from a different species or a cell derived from the same species. As used herein, “heterologous cell” means a cell derived from an organism of a different species from the recipient when a sheet-shaped cell culture is used for transplantation. For example, when the recipient is a human, cells derived from monkeys and pigs correspond to xenogeneic cells. "Allogeneic cell" means a cell derived from an organism of the same species as the recipient. For example, when the recipient is human, human cells correspond to cells derived from the same species. Allogeneic cells include autologous cells (also called autologous cells or autologous cells), that is, cells derived from the recipient and allogeneic non-autologous cells (also called allogeneic cells). Autologous cells are preferred in the present disclosure because rejection does not occur even when transplanted. However, it is also possible to use xenogeneic cells or allogeneic non-autologous cells. When xenogeneic cells or allogeneic non-autologous cells are used, immunosuppressive treatment may be necessary to suppress rejection. In this specification, cells other than autologous cells, that is, non-autologous cells of the same species as cells of xenogeneic origin may be collectively referred to as non-autologous cells. In one aspect of the present disclosure, the cells are autologous cells or allogeneic cells. In one aspect of the present disclosure, the cells are autologous cells (including autologous iPS cells). In another aspect of the present disclosure, the cells are allogeneic cells (including allogeneic iPS cells).
 本開示の移植片を構成する細胞は、多能性幹細胞から分化誘導された細胞であって、シート状細胞培養物などの移植片を形成し得るものであれば特に限定されず、例えば、接着細胞(付着性細胞)を含む。接着細胞は、例えば、接着性の体細胞(例えば、心筋細胞、線維芽細胞、上皮細胞、内皮細胞、肝細胞、膵細胞、腎細胞、副腎細胞、歯根膜細胞、歯肉細胞、骨膜細胞、皮膚細胞、滑膜細胞、軟骨細胞など)および幹細胞(例えば、筋芽細胞、心臓幹細胞などの組織幹細胞、間葉系幹細胞等)などを含む。体細胞は、幹細胞、特にiPS細胞から分化させたもの(iPS細胞由来接着細胞)であってもよい。シート状細胞培養物を構成する細胞の非限定例としては、例えば、iPS細胞由来の心筋細胞、線維芽細胞、上皮細胞、内皮細胞、肝細胞、膵細胞、腎細胞、副腎細胞、歯根膜細胞、歯肉細胞、骨膜細胞、皮膚細胞、滑膜細胞、軟骨細胞などが挙げられる。 Cells constituting the graft of the present disclosure are cells that have been induced to differentiate from pluripotent stem cells, and are not particularly limited as long as they can form a graft such as a sheet-shaped cell culture. Cells (adherent cells). Adherent cells include, for example, adherent somatic cells (eg, cardiomyocytes, fibroblasts, epithelial cells, endothelial cells, hepatocytes, pancreatic cells, kidney cells, adrenal cells, periodontal ligament cells, gingival cells, periosteal cells, skin Cells, synovial cells, chondrocytes, etc.) and stem cells (eg, tissue stem cells such as myoblasts, cardiac stem cells, mesenchymal stem cells, etc.). The somatic cells may be stem cells, particularly those differentiated from iPS cells (iPS cell-derived adherent cells). Non-limiting examples of cells constituting the sheet-shaped cell culture include, for example, iPS cell-derived cardiomyocytes, fibroblasts, epithelial cells, endothelial cells, hepatocytes, pancreatic cells, kidney cells, adrenal cells, periodontal ligament cells Gingival cells, periosteal cells, skin cells, synovial cells, chondrocytes and the like.
 移植片を構成する細胞は、移植片による治療が可能な任意の生物に由来し得る。かかる生物には、限定されずに、例えば、ヒト、非ヒト霊長類、イヌ、ネコ、ブタ、ウマ、ヤギ、ヒツジ、げっ歯目動物(例えば、マウス、ラット、ハムスター、モルモットなど)、ウサギなどが含まれる。また、移植片を構成する細胞の種類の数は特に限定されず、1種類のみの細胞で構成されていてもよいが、2種類以上の細胞を用いたものであってもよい。移植片を形成する細胞が2種類以上ある場合、最も多い細胞の含有比率(純度)は、移植片の形成終了時において、例えば50%以上、好ましくは60%以上、より好ましくは70%以上、さらに好ましくは75%以上であり得る。 細胞 The cells constituting the graft can be derived from any organism that can be treated with the graft. Such organisms include, without limitation, humans, non-human primates, dogs, cats, pigs, horses, goats, sheep, rodents (eg, mice, rats, hamsters, guinea pigs, etc.), rabbits, etc. Is included. The number of types of cells constituting the graft is not particularly limited, and may be composed of only one type of cell, or may be a type using two or more types of cells. When there are two or more types of cells forming the graft, the content ratio (purity) of the most abundant cells is, for example, 50% or more, preferably 60% or more, more preferably 70% or more at the end of the graft formation. More preferably, it can be 75% or more.
 培養基材は、細胞がその上で細胞培養物を形成し得るものであれば特に限定されず、例えば、種々の材質および/または形状の容器、容器中の固形もしくは半固形の表面などを含む。容器は、培養液などの液体を透過させない構造・材料が好ましい。かかる材料としては、限定することなく、例えば、ポリエチレン、ポリプロピレン、テフロン(登録商標)、ポリエチレンテレフタレート、ポリメチルメタクリレート、ナイロン6,6、ポリビニルアルコール、セルロース、シリコン、ポリスチレン、ガラス、ポリアクリルアミド、ポリジメチルアクリルアミド、金属(例えば、鉄、ステンレス、アルミニウム、銅、真鍮)等が挙げられる。また、容器は、少なくとも1つの平坦な面を有することが好ましい。かかる容器の例としては、限定することなく、例えば、細胞培養物の形成が可能な培養基材で構成された底面と、液体不透過性の側面とを備えた培養容器が挙げられる。かかる培養容器の特定の例としては、限定されずに、細胞培養皿、細胞培養ボトルなどが挙げられる。容器の底面は透明であっても不透明であってもよい。容器の底面が透明であると、容器の裏側から細胞の観察、計数などが可能となる。また、容器は、その内部に固形もしくは半固形の表面を有してもよい。固形の表面としては、上記のごとき種々の材料のプレートや容器などが、半固形の表面としては、ゲル、軟質のポリマーマトリックスなどが挙げられる。培養基材は、上記材料を用いて作製してもよいし、市販のものを利用してもよい。 The culture substrate is not particularly limited as long as cells can form a cell culture thereon, and includes, for example, containers of various materials and / or shapes, a solid or semi-solid surface in the container, and the like. . The container is preferably made of a structure / material that does not allow the passage of a liquid such as a culture solution. Examples of such a material include, but are not limited to, polyethylene, polypropylene, Teflon (registered trademark), polyethylene terephthalate, polymethyl methacrylate, nylon 6,6, polyvinyl alcohol, cellulose, silicon, polystyrene, glass, polyacrylamide, and polydimethyl. Acrylamide, metal (for example, iron, stainless steel, aluminum, copper, brass) and the like can be mentioned. Also, the container preferably has at least one flat surface. Examples of such a container include, without limitation, a culture container having a bottom surface formed of a culture substrate capable of forming a cell culture and a liquid impermeable side surface. Specific examples of such culture vessels include, but are not limited to, cell culture dishes, cell culture bottles, and the like. The bottom of the container may be transparent or opaque. If the bottom surface of the container is transparent, observation and counting of cells can be performed from the back side of the container. Further, the container may have a solid or semi-solid surface inside. Examples of the solid surface include plates and containers made of various materials as described above, and examples of the semi-solid surface include a gel and a soft polymer matrix. The culture substrate may be prepared using the above materials, or a commercially available substrate may be used.
 好ましい培養基材としては、限定することなく、例えば、シート状細胞培養物の形成に適した、接着性の表面を有する基材、スフェロイドの形成に適した、低接着性の表面を有する基材および/または均一なウェル状構造を有する基材などが挙げられる。具体的には、シート状細胞培養物の形成の場合であれば、例えば、コロナ放電処理したポリスチレン、コラーゲンゲルや親水性ポリマーなどの親水性化合物を該表面にコーティングした基材、さらには、コラーゲン、フィブロネクチン、ラミニン、ビトロネクチン、プロテオグリカン、グリコサミノグリカンなどの細胞外マトリックスや、カドヘリンファミリー、セレクチンファミリー、インテグリンファミリーなどの細胞接着因子などを表面にコーティングした基材などが挙げられる。また、かかる基材は市販されている(例えば、Corning(R) TC-Treated Culture Dish、Corningなど)。またスフェロイドの形成の場合であれば、例えば軟寒天、ポリ(N-イソプロピルアクリルアミド)(PIPAAm)をポリエチレングリコール(PEG)で架橋した温度応答性ゲル(市販名:メビオールゲル)、ポリメタクリル酸ヒドロキシエチル(ポリHEMA)、2-メタクリロイルオキシエチルホスホリスコリン(MPC)ポリマーなどのハイドロゲルなどの非細胞接着性化合物を表面にコーティングした基材および/または均一な凹凸構造を表面に有する基材などが挙げられる。かかる基材もまた市販されている(例えば、EZSPHERE(R)など)。培養基材は全体または部分が透明であっても不透明であってもよい。  Preferred culture substrates include, but are not limited to, for example, a substrate having an adhesive surface suitable for forming a sheet-shaped cell culture, and a substrate having a low adhesive surface suitable for forming a spheroid. And / or a substrate having a uniform well-like structure. Specifically, in the case of the formation of a sheet-shaped cell culture, for example, polystyrene subjected to corona discharge treatment, a substrate coated on its surface with a hydrophilic compound such as a collagen gel or a hydrophilic polymer, further, collagen And extracellular matrices such as fibronectin, laminin, vitronectin, proteoglycan, and glycosaminoglycan, and substrates coated on the surface with cell adhesion factors such as cadherin family, selectin family, and integrin family. Further, such substrates are commercially available (e.g., Corning (R) TC-Treated Culture Dish, Corning , etc.). In the case of spheroid formation, for example, temperature-responsive gel (commercial name: meviol gel) obtained by crosslinking soft agar, poly (N-isopropylacrylamide) (PIPAAm) with polyethylene glycol (PEG), polyhydroxyethyl methacrylate ( A substrate coated with a non-cell-adhesive compound such as a hydrogel such as poly (HEMA) or 2-methacryloyloxyethylphosphorhoscholine (MPC) polymer and / or a substrate having a uniform uneven structure on the surface. Can be Such substrates are also commercially available (e.g., EZSPHERE (R), etc.). The culture substrate may be entirely or partially transparent or opaque.
 培養基材は、刺激、例えば、温度や光に応答して物性が変化する材料で表面が被覆されていてもよい。かかる材料としては、限定されずに、例えば、(メタ)アクリルアミド化合物、N-アルキル置換(メタ)アクリルアミド誘導体(例えば、N-エチルアクリルアミド、N-n-プロピルアクリルアミド、N-n-プロピルメタクリルアミド、N-イソプロピルアクリルアミド、N-イソプロピルメタクリルアミド、N-シクロプロピルアクリルアミド、N-シクロプロピルメタクリルアミド、N-エトキシエチルアクリルアミド、N-エトキシエチルメタクリルアミド、N-テトラヒドロフルフリルアクリルアミド、N-テトラヒドロフルフリルメタクリルアミド等)、N,N-ジアルキル置換(メタ)アクリルアミド誘導体(例えば、N,N-ジメチル(メタ)アクリルアミド、N,N-エチルメチルアクリルアミド、N,N-ジエチルアクリルアミド等)、環状基を有する(メタ)アクリルアミド誘導体(例えば、1-(1-オキソ-2-プロペニル)-ピロリジン、1-(1-オキソ-2-プロペニル)-ピペリジン、4-(1-オキソ-2-プロペニル)-モルホリン、1-(1-オキソ-2-メチル-2-プロペニル)-ピロリジン、1-(1-オキソ-2-メチル-2-プロペニル)-ピペリジン、4-(1-オキソ-2-メチル-2-プロペニル)-モルホリン等)、またはビニルエーテル誘導体(例えば、メチルビニルエーテル)のホモポリマーまたはコポリマーからなる温度応答性材料、アゾベンゼン基を有する光吸収性高分子、トリフェニルメタンロイコハイドロオキシドのビニル誘導体とアクリルアミド系単量体との共重合体、および、スピロベンゾピランを含むN-イソプロピルアクリルアミドゲル等の光応答性材料などの公知のものを用いることができる(例えば、特開平2-211865、特開2003-33177参照)。これらの材料に所定の刺激を与えることによりその物性、例えば、親水性や疎水性を変化させ、同材料上に付着した細胞培養物の剥離を促進することができる。温度応答性材料で被覆された培養皿は市販されており(例えば、CellSeed Inc.のUpCell(R))、これらを本開示の製造方法に使用することができる。  The culture substrate may be coated on its surface with a material whose properties change in response to a stimulus, for example, temperature or light. Such materials include, but are not limited to, for example, (meth) acrylamide compounds, N-alkyl-substituted (meth) acrylamide derivatives (eg, N-ethylacrylamide, Nn-propylacrylamide, Nn-propylmethacrylamide, N-isopropylacrylamide, N-isopropylmethacrylamide, N-cyclopropylacrylamide, N-cyclopropylmethacrylamide, N-ethoxyethylacrylamide, N-ethoxyethylmethacrylamide, N-tetrahydrofurfurylacrylamide, N-tetrahydrofurfuryl methacryl Amide), N, N-dialkyl-substituted (meth) acrylamide derivatives (eg, N, N-dimethyl (meth) acrylamide, N, N-ethylmethylacrylamide, N, N-diethyl (Meth) acrylamide derivatives having a cyclic group (eg, 1- (1-oxo-2-propenyl) -pyrrolidine, 1- (1-oxo-2-propenyl) -piperidine, 4- (1-oxo) -2-propenyl) -morpholine, 1- (1-oxo-2-methyl-2-propenyl) -pyrrolidine, 1- (1-oxo-2-methyl-2-propenyl) -piperidine, 4- (1-oxo -2-methyl-2-propenyl) -morpholine or a homopolymer or copolymer of a vinyl ether derivative (eg, methyl vinyl ether), a light-absorbing polymer having an azobenzene group, triphenylmethane leucohydro A copolymer of a vinyl derivative of an oxide and an acrylamide monomer, and spirobenzopyra Can be used to include N- and isopropyl acrylamide gels known, such as photoresponsive materials (e.g., JP-A-2-211865, see JP-2003-33177). By applying a predetermined stimulus to these materials, their physical properties, for example, hydrophilicity or hydrophobicity, can be changed, and the detachment of the cell culture adhered on the materials can be promoted. Culture dishes coated with a temperature-responsive materials are commercially available (e.g., UpCell of CellSeed Inc. (R)), they can be used in the production method of the present disclosure.
 培養基材は、種々の形状であってもよい。また、その面積は特に限定されないが、例えば、約1cm~約200cm、約2cm~約100cm、約3cm~約50cmなどであってよい。例えば、培養基材として直径10cmの円形の培養皿が挙げられる。この場合、面積は56.7cmとなる。培養表面は平坦であってもよいし、凹凸構造を有していてもよい。凹凸構造を有する場合、均一な凹凸構造であることが好ましい。 The culture substrate may be in various shapes. The area is not particularly limited, but may be, for example, about 1 cm 2 to about 200 cm 2 , about 2 cm 2 to about 100 cm 2 , about 3 cm 2 to about 50 cm 2 , and the like. For example, a circular culture dish having a diameter of 10 cm is used as a culture substrate. In this case, the area is 56.7 cm 2 . The culture surface may be flat or may have an uneven structure. In the case of having an uneven structure, it is preferable to have a uniform uneven structure.
 本開示において、「人工多能性幹細胞」または「iPS細胞」は、当該技術分野で周知の用語であり、体細胞へ複数の遺伝子を導入することにより、三胚葉、すなわち内胚葉、中胚葉および外胚葉に属する全ての系列の細胞に分化することができる能力を獲得させた細胞を意味する。すなわち、人工多能性幹細胞またはiPS細胞とは、遺伝子を導入することにより誘導された、分化万能性および自己複製能を有する細胞である。iPS細胞を特定の細胞に分化誘導する際には、まずiPS細胞を浮遊培養して、上記三胚葉のいずれかの細胞の凝集体を形成し、その後凝集体を形成する細胞を目的とする特定の細胞に分化誘導させる。本開示において、iPS細胞は、任意の哺乳動物の細胞であり得るが、好ましくはヒト細胞から誘導されたiPS細胞である。 In the present disclosure, “inducible pluripotent stem cells” or “iPS cells” are terms well known in the art, and by introducing multiple genes into somatic cells, three germ layers, that is, endoderm, mesoderm and A cell that has acquired the ability to differentiate into all lineage cells belonging to the ectoderm. That is, induced pluripotent stem cells or iPS cells are cells having pluripotency and self-renewal ability induced by introducing a gene. When the iPS cells are induced to differentiate into specific cells, the iPS cells are first cultured in suspension to form aggregates of any of the above three germ layers, and then the cells that form the aggregates are identified. Cells are induced to differentiate. In the present disclosure, the iPS cells can be any mammalian cells, but are preferably iPS cells derived from human cells.
 本開示において、「iPS細胞由来の分化誘導細胞」は、iPS細胞から特定の種類の細胞に分化するように分化誘導処理された任意の細胞を意味する。分化誘導細胞の非限定例は、心筋細胞、骨格筋芽細胞などの筋肉系の細胞、ニューロン細胞、オリゴデンドロサイト、ドーパミン産生細胞などの神経系の細胞、網膜色素上皮細胞などの網膜細胞、血球細胞、骨髄細胞などの造血系の細胞、T細胞、NK細胞、NKT細胞、樹状細胞、B細胞などの免疫関連の細胞、肝細胞、膵β細胞、腎細胞などの臓器を構成する細胞、軟骨細胞、生殖細胞などの他、これらの細胞に分化する前駆細胞や体性幹細胞などを含む。かかる前駆細胞や体性幹細胞の典型例としては、例えば心筋細胞における間葉系幹細胞、多分化性心臓前駆細胞、単能性心臓前駆細胞、神経系の細胞における神経幹細胞、造血系の細胞や免疫関連の細胞における造血幹細胞およびリンパ系幹細胞などが挙げられる。iPS細胞の分化誘導は、既知の任意の手法を用いて行うことができる。例えば、iPS細胞から心筋細胞への分化誘導は、Miki et al., Cell Stem Cell 16, 699-711, June 4, 2015やWO2014/185358に記載の手法に基づいて行うことができる。 に お い て In the present disclosure, “differentiation-inducing cells derived from iPS cells” means any cells that have been subjected to differentiation-inducing treatment so as to differentiate from iPS cells into cells of a specific type. Non-limiting examples of differentiation-inducing cells include muscular cells such as cardiomyocytes and skeletal myoblasts, neuronal cells such as neuronal cells, oligodendrocytes and dopamine-producing cells, retinal cells such as retinal pigment epithelial cells, and blood cells. Cells, hematopoietic cells such as bone marrow cells, immune cells such as T cells, NK cells, NKT cells, dendritic cells, B cells, cells constituting organs such as hepatocytes, pancreatic β cells, kidney cells, In addition to chondrocytes, germ cells, and the like, progenitor cells and somatic stem cells that differentiate into these cells are included. Typical examples of such progenitor cells and somatic stem cells include, for example, mesenchymal stem cells in cardiomyocytes, pluripotent heart progenitor cells, unipotent heart progenitor cells, neural stem cells in nervous system cells, hematopoietic cells and immune cells. Related cells include hematopoietic stem cells and lymphoid stem cells. Induction of differentiation of iPS cells can be performed using any known technique. For example, differentiation induction from iPS cells to cardiomyocytes can be performed based on the method described in Miki et al., Cell Stem Cell 16, 699-711, June 4, 2015, and WO 2014/185358.
 また分化誘導細胞は、リプログラミングのための遺伝子以外の任意の有用な遺伝子が導入されたiPS細胞から誘導された細胞であってもよい。かかる細胞の非限定例としては、例えば、Themeli M. et al. Nature Biotechnology, vol. 31, no. 10, pp. 928-933, 2013に記載のキメラ抗原受容体の遺伝子が導入されたiPS細胞から誘導されるT細胞などが挙げられる。また、iPS細胞から分化誘導された後、任意の有用な遺伝子が導入された細胞もまた、本発明の分化誘導細胞に包含される。 The differentiation-inducing cell may be a cell derived from an iPS cell into which any useful gene other than a gene for reprogramming has been introduced. Non-limiting examples of such cells include, for example, iPS cells into which the gene for the chimeric antigen receptor described in Themeli M. et al. Nature Biotechnology, vol. 31, vol. 10, pp. 928-933, 2013 has been introduced. And T cells derived therefrom. In addition, cells into which any useful gene has been introduced after differentiation induction from iPS cells are also included in the differentiation-inducing cells of the present invention.
 以下に所望の細胞がiPS細胞から分化誘導された心筋細胞であり、移植片がシート状細胞培養物である場合を例として、本発明を詳述する。
 本開示の一側面は、iPS細胞から分化誘導された心筋細胞を含む高品質なシート状細胞培養物を製造する方法に関する。本開示の方法は、以下の工程(a)および(b)を含む:
(a)iPS細胞から分化誘導された心筋細胞を含む細胞集団を、培養基材上に播種する工程、および
(b)播種した細胞集団を、血小板溶解物を含むシート化媒体でシート化培養する工程。
Hereinafter, the present invention will be described in detail by taking as an example a case where the desired cells are cardiomyocytes differentiated from iPS cells and the transplant is a sheet-shaped cell culture.
One aspect of the present disclosure relates to a method for producing a high-quality sheet-shaped cell culture containing cardiomyocytes differentiated from iPS cells. The method of the present disclosure includes the following steps (a) and (b):
(A) a step of seeding a cell population containing cardiomyocytes differentiated from iPS cells on a culture substrate; and (b) a sheet culture of the seeded cell population with a sheeting medium containing a platelet lysate. Process.
 本開示において、「心筋細胞」とは、心筋細胞の特徴を有する細胞を意味する。心筋細胞の特徴としては、限定されずに、例えば、心筋細胞マーカーの発現、自律的拍動の存在などが挙げられる。心筋細胞マーカーの非限定例としては、例えば、c-TNT(cardiac troponin T)、CD172a(別名SIRPAまたはSHPS-1)、KDR(別名CD309、FLK1またはVEGFR2)、PDGFRA、EMILIN2、VCAMなどが挙げられる。一態様において、iPS細胞由来の心筋細胞は、c-TNT陽性かつ/またはCD172a陽性である。 に お い て In the present disclosure, “cardiomyocytes” means cells having characteristics of cardiomyocytes. The characteristics of the cardiomyocyte include, but are not limited to, for example, the expression of a cardiomyocyte marker, the presence of an autonomous beat, and the like. Non-limiting examples of cardiomyocyte markers include, for example, c-TNT (cardiac troponin T), CD172a (alias SIRPA or SHPS-1), KDR (alias CD309, FLK1 or VEGFR2), PDGFRA, EMILIN2, VCAM and the like. . In one aspect, the iPS cell-derived cardiomyocytes are c-TNT positive and / or CD172a positive.
 工程(a)において、培養基材への播種は、例えば、細胞をシート化媒体に懸濁した細胞懸濁液を、培養基材を備えた培養容器に注入することなどにより行ってもよい。細胞懸濁液の注入には、スポイトやピペットなど、細胞懸濁液の注入操作に適した器具を用いることができる。細胞の播種密度は、シート状細胞培養物を形成し得る密度で行われ、かかる密度は所望の細胞により異なり得るが、当業者であれば当該技術分野において公知の手法などから適切な密度を選択することができる。例えば心筋細胞を含むシート状細胞培養物である場合、例えば2.0×10個/cm以上などであり得るが、より高密度で播種してもよい。 In the step (a), the seeding on the culture substrate may be performed, for example, by injecting a cell suspension in which cells are suspended in a sheeting medium into a culture container provided with the culture substrate. For injection of the cell suspension, a device suitable for the operation of injecting the cell suspension, such as a dropper or pipette, can be used. The seeding density of the cells is determined at a density capable of forming a sheet-shaped cell culture, and the density may vary depending on the desired cells. However, those skilled in the art can select an appropriate density from techniques known in the art. can do. For example, in the case of a sheet-shaped cell culture containing cardiomyocytes, it may be, for example, 2.0 × 10 5 cells / cm 2 or more, but may be seeded at a higher density.
 より高密度の例としては、例えばコンフルエントに達する密度、すなわち播種した際に細胞が培養容器の接着表面一面を覆うことが想定される程度の密度、例えば、播種した際に、細胞が互いに接触することが想定される程度の密度、接触阻害が発生する密度、または接触阻害により細胞の増殖を実質的に停止する密度であり得る。播種密度の上限は、特に制限されないが、密度が過度に高い場合には、死滅する細胞が多くなり、非効率となる。本開示の一態様において、播種密度は、例えば約1.0×10個/cm~約1.0×10個/cm、約1.0×10個/cm~約5.0×10個/cm、約1.0×10個/cm~約3.0×10個/cm、約1.5×10個/cm~約1.0×10個/cm、約1.5×10個/cm~約5.0×10個/cm、約1.5×10個/cm~約3.0×10個/cm、約2.0×10個/cm~約1.0×10個/cm、約2.0×10個/cm~約5.0×10個/cm、約2.0×10個/cm~約3.0×10個/cmなどであり得る。好ましい一態様において、播種密度は、約1.76×10個/cm~約2.33×10個/cmである。 Examples of higher densities include, for example, densities that reach confluence, i.e., densities at which cells are expected to cover the entire adhesive surface of the culture vessel upon seeding, e.g., upon seeding, cells contact each other , A density at which contact inhibition occurs, or a density at which cell growth is substantially stopped by contact inhibition. The upper limit of the seeding density is not particularly limited. However, if the seeding density is excessively high, the number of dead cells increases, resulting in inefficiency. In one aspect of the present disclosure, the seeding density is, for example, from about 1.0 × 10 6 / cm 2 to about 1.0 × 10 7 / cm 2 , from about 1.0 × 10 6 / cm 2 to about 5 0.0 × 10 6 / cm 2 , about 1.0 × 10 6 / cm 2 to about 3.0 × 10 6 / cm 2 , about 1.5 × 10 6 / cm 2 to about 1.0 × 10 7 pieces / cm 2 , about 1.5 × 10 6 pieces / cm 2 to about 5.0 × 10 6 pieces / cm 2 , about 1.5 × 10 6 pieces / cm 2 to about 3.0 × 10 6 pieces / cm 2 , about 2.0 × 10 6 pieces / cm 2 to about 1.0 × 10 7 pieces / cm 2 , about 2.0 × 10 6 pieces / cm 2 to about 5.0 × 10 6 pieces / Cm 2 , about 2.0 × 10 6 / cm 2 to about 3.0 × 10 6 / cm 2 , and the like. In one preferred embodiment, the seeding density is from about 1.76 × 10 6 / cm 2 to about 2.33 × 10 6 / cm 2 .
 播種される培養基材は、上記で詳述したとおりであるが、好ましい一態様において、細胞外マトリクスや細胞接着因子などの細胞接着性成分を表面にコーティングした培養基材である。細胞接着性成分としては、これに限定するものではないが、例えばコラーゲン、フィブロネクチン、ラミニン、ビトロネクチン、プロテオグリカン、グリコサミノグリカンなどの細胞外マトリックス、カドヘリンファミリー、セレクチンファミリー、インテグリンファミリーなどの細胞接着因子などが挙げられるほか、これらの改変物、例えばラミニン511(ラミニンの改変物)、VTN-N(ビトロネクチンの改変物)、レトロネクチン(R)(フィブロネクチンの改変物)、であってもよい。 The culture substrate to be seeded is as described in detail above, but in a preferred embodiment, is a culture substrate having a surface coated with a cell adhesive component such as an extracellular matrix or a cell adhesion factor. Cell adhesion components include, but are not limited to, for example, extracellular matrices such as collagen, fibronectin, laminin, vitronectin, proteoglycans, glycosaminoglycans, cadherin family, selectin family, cell adhesion factors such as integrin family And modifications thereof, for example, laminin 511 (a modification of laminin), VTN-N (a modification of vitronectin), and RetroNectin (R) (a modification of fibronectin).
 播種される細胞集団は、iPS細胞から分化誘導された心筋細胞を含んでいれば、他の細胞を含んでいてもよい。一般にiPS細胞から心筋細胞へ分化誘導して得られる細胞集団には、心筋細胞の他、例えば線維芽細胞や血管内皮細胞などが含まれ得る。細胞集団は、例えば上記Miki et al., Cell Stem Cell 16, 699-711, June 4, 2015やWO2014/185358に記載の手法などを用いてiPS細胞から分化誘導して得られた細胞集団をそのまま用いてもよいし、凍結保存やプレ培養などを実施した後に用いてもよい。好ましい一態様において、播種される細胞集団は、iPS細胞から分化誘導後、培養基材上(好ましくは平面状の培養基材上)に播種して接着培養を行い、その後回収された細胞集団である。かかる接着培養の前または後に、凍結保存および解凍を実施してもよい。接着培養を行うことにより、その後の移植片の形成において、高品質な移植片の形成を、高確率で達成することが可能となる。一態様において、形成された移植片には心筋細胞のほかにのほかに血管内皮細胞、細胞壁、線維芽細胞を含んでもよい。本開示の移植片に含まれる細胞の構成比は、例えば、心筋細胞約30~70%、血管内皮細胞0.1%~約20%、および壁細胞約1%~約40%であってもよい。
 かかる接着培養ステップにおいて、培養条件などは、通常の接着培養を行う場合の条件に準じてよい。例えば、市販の接着培養用培養容器を用いて、37℃、5%CO条件下での培養などであってよい。細胞の播種密度は、細胞同士の接着および/または細胞と培養基材との接着の形成を妨げない密度であればいかなる密度であってもよく、例えばサブコンフルエントな密度であってもよいし、コンフルエントに達する密度またはそれ以上であってもよい。培養時間は、細胞同士の接着および/または細胞と培養基材との接着が形成される程度の時間であればよく、具体的には例えば2~168時間、2~144時間、2~120時間、2~96時間、2~72時間、2~48時間、2~24時間、2~12時間、2~6時間、2~4時間程度であればよい。
The seeded cell population may contain other cells as long as it contains cardiomyocytes differentiated from iPS cells. In general, a cell population obtained by inducing differentiation from iPS cells into cardiomyocytes may include, for example, fibroblasts and vascular endothelial cells in addition to cardiomyocytes. As the cell population, for example, a cell population obtained by inducing differentiation from iPS cells using the method described in Miki et al., Cell Stem Cell 16, 699-711, June 4, 2015 or WO 2014/185358, or the like is used as it is. It may be used, or may be used after carrying out cryopreservation or pre-culture. In a preferred embodiment, the cell population to be seeded is induced to differentiate from iPS cells, and then inoculated on a culture substrate (preferably on a planar culture substrate) to perform adherent culture. is there. Cryopreservation and thawing may be performed before or after such adherent culture. By performing the adherent culture, it is possible to achieve the formation of a high-quality graft at a high probability in the subsequent formation of the graft. In one embodiment, the formed graft may include vascular endothelial cells, cell walls, fibroblasts in addition to cardiomyocytes. The percentage of cells contained in the graft of the present disclosure may be, for example, about 30-70% of cardiomyocytes, 0.1% to about 20% of vascular endothelial cells, and about 1% to about 40% of mural cells. Good.
In such an adhesion culture step, the culture conditions and the like may be in accordance with the conditions for performing ordinary adhesion culture. For example, the culture may be performed at 37 ° C. under 5% CO 2 using a commercially available culture vessel for adhesion culture. The seeding density of the cells may be any density as long as the density does not prevent adhesion between cells and / or formation of adhesion between the cells and the culture substrate, for example, a subconfluent density, The density may be confluent or higher. The cultivation time may be a time period such that adhesion between cells and / or adhesion between cells and a culture substrate is formed, and specifically, for example, 2 to 168 hours, 2 to 144 hours, 2 to 120 hours It may be about 2 to 96 hours, 2 to 72 hours, 2 to 48 hours, 2 to 24 hours, 2 to 12 hours, 2 to 6 hours, 2 to 4 hours.
 工程(b)において、播種した細胞をシート化培養する。本明細書においては、播種した細胞を移植片として形成するための培養を「移植片形成培養」と称し、移植片がシート状細胞培養物であり、播種した細胞をシート化するための培養を特に、「シート化培養」と称する。播種した細胞のシート化は、既知の任意の手法および条件で行うことができる。かかる手法の非限定例は、例えば、特開2010-081829、特開2010-226991、特開2011-110368、特開2011-172925、WO 2014/185517などに記載されている。細胞のシート化は、細胞同士が接着分子や、細胞外マトリックスなどの細胞間接着機構を介して互いに接着することにより達成されると考えられている。したがって、播種した細胞をシート化するステップは、例えば、細胞を、細胞間接着を形成する条件下で培養することにより達成することができる。かかる条件は、細胞間接着を形成することができればいかなるものであってもよいが、通常は一般的な細胞培養条件と同様の条件であれば細胞間接着を形成することができる。かかる条件としては、例えば、約37℃、5%COでの培養が挙げられる。また、培養は通常の圧力下(大気圧下、非加圧下)で行うことができる。培養は任意の大きさおよび形状の容器で行うことができる。シート状細胞培養物の大きさや形状は、培養容器の細胞付着面の大きさ・形状を調整すること、または、培養容器の細胞付着面に、所望の大きさ・形状の型枠を設置し、その内部で細胞を培養することなどにより任意に調節することができる。 In the step (b), the seeded cells are cultured on a sheet. In the present specification, a culture for forming the seeded cells as a graft is referred to as a “graft formation culture”, and the graft is a sheet-shaped cell culture, and the culture for forming the seeded cells into a sheet is performed. In particular, it is referred to as “sheet culture”. Sheeting of the seeded cells can be performed by any known method and conditions. Non-limiting examples of such techniques are described in, for example, JP-A-2010-081829, JP-A-2010-226991, JP-A-2011-110368, JP-A-2011-172925, WO 2014/185517, and the like. It is believed that cell sheeting is achieved by cells adhering to each other via intercellular adhesion mechanisms such as adhesion molecules and extracellular matrix. Therefore, the step of forming the seeded cells into a sheet can be achieved, for example, by culturing the cells under conditions that form intercellular adhesion. Such conditions may be any as long as they can form cell-cell adhesion, but usually, cell-cell adhesion can be formed under the same conditions as general cell culture conditions. Such conditions include, for example, culture at about 37 ° C., 5% CO 2 . The cultivation can be performed under normal pressure (atmospheric pressure, non-pressurized). The culture can be performed in a container of any size and shape. The size and shape of the sheet-shaped cell culture can be adjusted by adjusting the size and shape of the cell attachment surface of the culture container, or by installing a mold of the desired size and shape on the cell attachment surface of the culture container, It can be adjusted arbitrarily by culturing the cells therein.
 シート化培養の時間は、播種する細胞の種類や細胞密度により異なり得る。例えばiPS細胞から心筋細胞を調製してシート化する場合、例えば約2.1×10個/cmなどの密度で播種し、4日以上培養することによりシート化を行ってよい。また、播種密度をコンフルエントに達する密度、すなわちより高密度で播種する場合、シート化培養の期間を短縮することができ、培養時間は2~4日、より好ましくは2~3日であってよい。 The time of sheeting culture may vary depending on the type of seeded cells and cell density. For example, when cardiomyocytes are prepared from iPS cells and formed into sheets, the sheets may be seeded at a density of, for example, about 2.1 × 10 5 cells / cm 2 and cultured for 4 days or more to form sheets. When the seeding density reaches a confluent density, that is, when the seeding is performed at a higher density, the period of sheet culture can be shortened, and the culture time may be 2 to 4 days, more preferably 2 to 3 days. .
 移植片形成(例えばシート化)に用いる媒体(移植片形成媒体、移植片形成がシート化である場合は特にシート化媒体と称する場合もある)としては、細胞の移植片形成を可能にするものであれば特に限定されず、例えば、生理食塩水、種々の生理緩衝液(例えば、PBS、HBSS等)、種々の細胞培養用の基礎培地をベースにしたものなどを使用してもよい。かかる基礎培地には、限定されずに、例えば、DMEM、MEM、F12、DME、RPMI1640、MCDB(MCDB102、104、107、120、131、153、199など)、L15、SkBM、RITC80-7、DMEM/F12などが含まれる。これらの基礎培地の多くは市販されており、その組成も公知となっている。基礎培地は、標準的な組成のまま(例えば、市販されたままの状態で)用いてもよいし、細胞種や細胞条件に応じてその組成を適宜変更してもよい。したがって、本発明に用いる基礎培地は、公知の組成のものに限定されず、1または2以上の成分が追加、除去、増量もしくは減量されたものを含む。移植片形成媒体は、通常血清(例えば、ウシ胎仔血清などのウシ血清、ウマ血清、ヒト血清等)、種々の成長因子(例えば、FGF、EGF、VEGF、HGF等)などの添加物を含んでもよいが、移植片をゼノフリー条件下で製造する場合、特にウシ血清、ウマ血清など、移植片に含まれる細胞が由来する種と異種の血清を含まないことが好ましい。本開示は、血清や成長因子に代えてまたは加えて、血小板溶解物を含む移植片形成媒体で移植片形成培養を行うことを特徴とする。好ましい一態様において、移植片形成媒体は血小板溶解物を含むが、血清を含まない。 As a medium used for graft formation (for example, sheeting) (a graft forming medium, sometimes referred to as a sheeting medium particularly when the graft formation is a sheeting), a medium capable of forming a cell graft is used. There is no particular limitation so long as it is, for example, physiological saline, various physiological buffers (for example, PBS, HBSS, etc.), or those based on various cell culture base media. Examples of such a basal medium include, but are not limited to, DMEM, MEM, F12, DME, RPMI1640, MCDB (MCDB102, 104, 107, 120, 131, 153, 199, etc.), L15, SkBM, RITC80-7, DMEM / F12 and the like. Many of these basal media are commercially available and their compositions are also known. The basal medium may be used with its standard composition (for example, as it is commercially available), or its composition may be appropriately changed depending on the cell type and cell conditions. Therefore, the basal medium used in the present invention is not limited to those having a known composition, and includes those in which one or more components have been added, removed, increased or reduced in weight. The graft forming medium may contain additives such as normal serum (eg, bovine serum such as fetal bovine serum, horse serum, human serum, etc.) and various growth factors (eg, FGF, EGF, VEGF, HGF, etc.). Preferably, when the transplant is produced under xeno-free conditions, it is preferable that the transplant contains no serum different from the species from which the cells contained in the transplant are derived, such as bovine serum and horse serum. The present disclosure is characterized in that a graft forming culture is performed using a graft forming medium containing a platelet lysate instead of or in addition to serum or a growth factor. In one preferred embodiment, the graft forming medium comprises platelet lysate but does not contain serum.
 本開示において、「血小板溶解物」(Platelet lysate:PL)は、血小板に対して凍結融解を繰り返すことにより得られる、成長因子等を豊富に含む組成物をいう。血小板溶解物は、近年では、間葉系幹細胞の増殖を促進することなどが知られている。本発明者らは、iPS細胞から誘導された心筋細胞を含むシート状細胞培養物の製造において、シート化媒体に血小板溶解物を含有せしめることにより、従来よりも早い培養日数で、強い自律拍動が観察されることを初めて見出した。 に お い て In the present disclosure, “platelet lysate (PL)” refers to a composition rich in growth factors and the like, obtained by repeatedly freezing and thawing platelets. In recent years, platelet lysates have been known to promote the growth of mesenchymal stem cells. In the production of a sheet-shaped cell culture containing cardiomyocytes derived from iPS cells, the present inventors have added a platelet lysate to a sheeting medium, thereby achieving a strong autonomic pulsation in a shorter culture period than before. Was observed for the first time.
 血小板溶解物は、細胞培養用の培地添加物として市販されており、当該技術分野において公知である。血小板溶解物は、例えば特表2014-533715、Bieback et al., STEM CELLS, 2009;27:2331-2341などに記載の方法などにより調製可能である。
 具体的な調製方法としては、例えば、血小板の集団を溶解させ、そこから血小板の粒子や膜などの夾雑物を除去して上清を得るなどの手段により調製できる。血小板の溶解は、化学的手段(例えばCaClの使用など)、浸透圧的手段(例えば蒸留水の使用など)、凍結融解手段、機械的破壊手段などの工程を介して達成することができる。夾雑物の除去は、遠心分離やろ過などの方法により達成することができる。
Platelet lysates are commercially available as media additives for cell culture and are known in the art. The platelet lysate can be prepared, for example, by the method described in JP-T-2014-533715, Bieback et al., STEM CELLS, 2009; 27: 2331-2341.
As a specific preparation method, it can be prepared by, for example, dissolving a platelet population and removing contaminants such as platelet particles and a membrane therefrom to obtain a supernatant. Lysis of platelets can be achieved through steps such as chemical means (eg, using CaCl 2 ), osmotic means (eg, using distilled water), freeze-thaw means, mechanical disruption means, and the like. Removal of contaminants can be achieved by a method such as centrifugation or filtration.
 移植片形成媒体中に含まれる血小板溶解物の濃度は、当該技術分野において通常用いられる程度であればよく、例えば1%、2.5%、5%、10%、15%、20%などであってよい。好ましい一態様において、血小板溶解物は、移植片形成媒体中に1%~20%、より好ましくは2%~10%、さらに好ましくは2.5%~10%程度含有される。 The concentration of the platelet lysate contained in the graft forming medium may be a level generally used in the art, such as 1%, 2.5%, 5%, 10%, 15%, 20%, and the like. May be. In a preferred embodiment, the platelet lysate is contained in the graft forming medium in an amount of about 1% to 20%, more preferably about 2% to 10%, and still more preferably about 2.5% to 10%.
 移植片形成媒体は、移植片形成培養中に適宜入れ替えてよい。また、移植片形成の進行に合わせて媒体の組成を変化させてもよい。本発明者らは、iPS細胞から誘導された心筋細胞を含むシート状細胞培養物の製造において、Rhoキナーゼ(ROCK)阻害剤を添加したシート化媒体を、シート化培養1日目の媒体として用いることにより、効果的にシート状細胞培養物が形成されることを新たに見出した。したがって本開示の好適な一態様において、1日目のシート化培養に用いるシート化媒体は、Rhoキナーゼ阻害剤を含む。かかる態様においては、2日目以降のシート化媒体にはRhoキナーゼ阻害剤を含んでも含まなくてもよいが、好ましくはRhoキナーゼ阻害剤を含まない。 The graft forming medium may be appropriately replaced during the graft forming culture. Further, the composition of the medium may be changed in accordance with the progress of the graft formation. The present inventors use a sheeting medium to which a Rho kinase (ROCK) inhibitor is added as a medium on the first day of sheeting culture in the production of a sheet-shaped cell culture containing cardiomyocytes derived from iPS cells. As a result, it was newly found that a sheet-shaped cell culture was effectively formed. Thus, in one preferred aspect of the present disclosure, the sheeting medium used for day 1 sheeting culture comprises a Rho kinase inhibitor. In such an embodiment, the sheeting medium after the second day may or may not contain a Rho kinase inhibitor, but preferably does not contain a Rho kinase inhibitor.
 シート化媒体は、さらに細胞接着性成分を含んでよい。細胞接着性成分については上記で詳述したとおりである。シート化媒体に細胞接着性成分が含まれる場合、培養基材は細胞接着性成分でコーティングされていてもコーティングされていなくてもよい。培養基材が細胞接着性成分でコーティングされている場合、シート化媒体に含まれる細胞接着性成分は、培養基材をコーティングしている細胞接着性成分と同一であってもよいし異なっていてもよいが、好ましくは同一の細胞接着性成分である。 The sheeting medium may further contain a cell adhesive component. The cell adhesive component is as described in detail above. When the sheet-forming medium contains a cell adhesive component, the culture substrate may or may not be coated with the cell adhesive component. When the culture substrate is coated with the cell adhesive component, the cell adhesive component contained in the sheeting medium may be the same as or different from the cell adhesive component coating the culture substrate. However, they are preferably the same cell adhesive component.
 移植片形成媒体に含まれる細胞接着性成分の濃度は、含まれる細胞接着性成分の種類や移植片形成する細胞の状態などにより異なり得る。例えば、バイアビリティの低い、すなわち活性が弱い細胞を用いている場合、細胞接着性成分の含有量は少ない方がよい。シート化媒体に含まれる細胞接着性成分の濃度は、培養基材のコーティング剤として同じ細胞接着性成分を用いる場合に使用する濃度を基準(100%)として、約0.1%、約0.5%、約1%、約5%、約10%、約20%、約25%、約50%、約75%、約100%などであってよい。したがって好ましい一態様において、シート化媒体に含まれる細胞接着性成分の濃度範囲は、培養基材のコーティング剤として同じ細胞接着性成分を用いる場合に使用する濃度を基準(100%)として、約0.1%~約100%、約0.1%~約100%、約0.1%~約50%、約0.1%~約25%、約0.1%~約20%、約0.1%~約10%、約1%~約100%、約1%~約100%、約0.5%~約100%、約0.5%~約100%、約0.5%~約50%、約0.5%~約25%、約0.5%~約20%、約0.5%~約10%、約1%~約50%、約1%~約25%、約1%~約20%、約1%~約10%、約0.5%~約100%、約5%~約100%、約5%~約50%、約5%~約25%、約5%~約20%、約5%~約10%などであってよい。 濃度 The concentration of the cell-adhesive component contained in the graft-forming medium may vary depending on the type of the cell-adhesive component contained, the state of the cells forming the graft, and the like. For example, when cells having low viability, that is, cells having weak activity, are used, the content of the cell adhesive component is preferably small. The concentration of the cell adhesive component contained in the sheeting medium is about 0.1% and about 0.1% based on the concentration (100%) used when the same cell adhesive component is used as a coating agent for the culture substrate. It may be 5%, about 1%, about 5%, about 10%, about 20%, about 25%, about 50%, about 75%, about 100%, etc. Therefore, in a preferred embodiment, the concentration range of the cell adhesive component contained in the sheeting medium is about 0% based on the concentration (100%) used when the same cell adhesive component is used as a coating agent for the culture substrate. 0.1% to about 100%, about 0.1% to about 100%, about 0.1% to about 50%, about 0.1% to about 25%, about 0.1% to about 20%, about 0% 0.1% to about 10%, about 1% to about 100%, about 1% to about 100%, about 0.5% to about 100%, about 0.5% to about 100%, about 0.5% to About 50%, about 0.5% to about 25%, about 0.5% to about 20%, about 0.5% to about 10%, about 1% to about 50%, about 1% to about 25%, About 1% to about 20%, about 1% to about 10%, about 0.5% to about 100%, about 5% to about 100%, about 5% to about 50%, about 5% to about 25%, About 5% to about 20%, May the like 5% to about 10%.
 本開示の別の側面は、iPS細胞から分化誘導した心筋細胞を含む移植片において心筋細胞の拍動開始を早める方法であって、前記心筋細胞を含む細胞集団を、血小板溶解物を含む移植片形成媒体中で移植片形成培養することを含む、前記方法に関する。本発明者らは、iPS細胞から分化誘導した心筋細胞を含む移植片を形成する際に移植片形成媒体に血小板溶解物を添加することにより、無血清媒体での移植片形成を行う場合と比較して心筋細胞の拍動開始が早まることを見出した。これにより、ゼノフリー環境において簡便に、かつ高品質な移植片を形成することが可能となる。 Another aspect of the present disclosure is a method of hastening the onset of pulsation of cardiomyocytes in a graft containing cardiomyocytes differentiated from iPS cells, comprising: The present invention relates to the above method, comprising culturing the explant in a formation medium. The present inventors compared platelet formation with a serum-free medium by adding a platelet lysate to a graft formation medium when forming a graft containing cardiomyocytes differentiated from iPS cells. It was found that the onset of pulsation of cardiomyocytes was accelerated. This makes it possible to easily form a high-quality implant in a xeno-free environment.
 本開示の別の側面は、本開示の方法により製造された移植片の有効量を、それを必要とする対象に適用することを含む、前記対象における疾患を処置する方法に関する。処置の対象となる疾患は、上記したとおりである。 別 Another aspect of the present disclosure relates to a method of treating a disease in a subject, comprising applying an effective amount of an implant produced by the method of the present disclosure to the subject in need thereof. The disease to be treated is as described above.
 本開示において、用語「処置」は、疾患の治癒、一時的寛解または予防などを目的とする医学的に許容される全ての種類の予防的および/または治療的介入を包含するものとする。例えば、「処置」の用語は、組織の異常に関連する疾患の進行の遅延または停止、病変の退縮または消失、当該疾患発症の予防または再発の防止などを含む、種々の目的の医学的に許容される介入を包含する。 に お い て In the present disclosure, the term “treatment” is intended to include all types of medically acceptable prophylactic and / or therapeutic interventions, such as for the cure, temporary remission or prevention of disease. For example, the term "treatment" includes medically acceptable treatments for a variety of purposes, including slowing or stopping the progression of a disease associated with tissue abnormalities, regressing or eliminating lesions, preventing the onset of the disease or preventing its recurrence, and the like. Involve interventions.
 本開示の処置方法においては、移植片の生存性、生着性および/または機能などを高める成分や、対象疾患の処置に有用な他の有効成分などを、本開示の移植片等と併用することができる。 In the treatment method of the present disclosure, a component that enhances the survival, engraftment, and / or function of a graft, and other active components that are useful for treating a target disease are used in combination with the graft or the like of the present disclosure. be able to.
 本開示の処置方法は、本開示の製造方法に従って、本開示の移植片を製造するステップをさらに含んでもよい。本開示の処置方法は、移植片を製造するステップの前に、対象から移植片を製造するための細胞(例えば、皮膚細胞、血球等の、iPS細胞へと誘導するための体細胞)または細胞の供給源となる組織(例えば、皮膚組織、血液等の、iPS細胞へと誘導するための体細胞を含む組織)を採取するステップをさらに含んでもよい。一態様において、細胞または細胞の供給源となる組織を採取する対象は、細胞培養物、組成物、または移植片等の投与を受ける対象と同一の個体である。別の態様において、細胞または細胞の供給源となる組織を採取する対象は、細胞培養物、組成物、または移植片等の投与を受ける対象とは同種の別個体である。別の態様において、細胞または細胞の供給源となる組織を採取する対象は、移植片等の投与を受ける対象とは異種の個体である。 処置 The treatment method of the present disclosure may further include a step of manufacturing the implant of the present disclosure according to the manufacturing method of the present disclosure. The method of treatment of the present disclosure may comprise, prior to the step of producing the graft, cells (eg, somatic cells for inducing iPS cells, such as skin cells, blood cells, etc.) or cells for producing the graft from the subject. The method may further include a step of collecting a tissue (for example, a tissue containing a somatic cell for inducing iPS cells, such as skin tissue or blood) serving as a source of the protein. In one embodiment, the subject from whom the cells or tissue from which the cells are to be sourced is harvested is the same individual as the subject to whom a cell culture, composition, or explant is administered. In another embodiment, the subject from whom the cells or tissue from which the cells are to be sourced is harvested is a homologous distinct body from the subject to be administered, such as a cell culture, composition, or implant. In another embodiment, the subject from which the cells or the tissue from which the cells are sourced is harvested is an individual that is heterogeneous to the subject receiving the administration, such as a graft.
 本開示において、有効量とは、例えば、疾患の発症や再発を抑制し、症状を軽減し、または進行を遅延もしくは停止し得る量(例えば、移植片のサイズ、重量、数等)であり、好ましくは、当該疾患の発症および再発を予防し、または当該疾患を治癒する量である。また、投与による利益を超える悪影響が生じない量が好ましい。かかる量は、例えば、マウス、ラット、イヌまたはブタなどの実験動物や疾患モデル動物における試験などにより適宜決定することができ、このような試験法は当業者によく知られている。また、処置の対象となる組織病変の大きさは、有効量決定のための重要な指標となり得る。 In the present disclosure, an effective amount is, for example, an amount capable of suppressing the onset or recurrence of a disease, reducing symptoms, or delaying or stopping the progress (eg, size, weight, number, etc. of a graft), Preferably, it is an amount that prevents the onset and recurrence of the disease or cures the disease. Also preferred is an amount that does not cause adverse effects beyond the benefit of administration. Such an amount can be appropriately determined, for example, by a test in a laboratory animal such as a mouse, a rat, a dog or a pig, or a disease model animal, and such a test method is well known to those skilled in the art. In addition, the size of a tissue lesion to be treated can be an important index for determining an effective amount.
 投与方法としては、例えば、静脈投与、筋肉内投与、骨内投与、髄腔内投与、組織への直接的な適用などが挙げられる。投与頻度は、典型的には1回の処置につき1回であるが、所望の効果が得られない場合には、複数回投与することも可能である。組織に適用する際、本発明の細胞培養物、組成物、またはシート状細胞培養物等を対象の組織に縫合糸やステープルなどの係止手段により固定してもよい。 Examples of the administration method include intravenous administration, intramuscular administration, intraosseous administration, intrathecal administration, and direct application to tissues. The frequency of administration is typically once per treatment, but multiple administrations are possible if the desired effect is not obtained. When applied to a tissue, the cell culture, the composition, the sheet-shaped cell culture, or the like of the present invention may be fixed to a target tissue by a locking means such as a suture or staple.
 本発明を以下の例を参照してより詳細に説明するが、これらは本発明の特定の具体例を示すものであり、本発明はこれらに限定されるものではない。 The present invention will be described in more detail with reference to the following examples, which show specific embodiments of the present invention, and the present invention is not limited thereto.
 以下の実施例において、多能性幹細胞として、京都大学iPS細胞研究所(CiRA)で樹立された臨床用ヒトiPS細胞を用いた。M. Nakagawa et al., Scientific Reports, 4:3594 (2014)を参考に、ヒトiPS細胞をフィーダーフリー法で維持した。ついで、Miki et al., Cell Stem Cell 16, 699-711, June 4, 2015やWO2014/185358およびWO2017/038562の記載を参考にして、ヒトiPS細胞を心筋細胞へと分化誘導して胚様体を得た。具体的には、フィーダー細胞を含まない培養液で維持培養したヒトiPS細胞を、EZ Sphere(旭硝子)上で10μMのY27632(和光純薬)を含有するStemFit AK03培地(味の素)中で1日培養し、得られた胚様体をアクチビンA、骨形成タンパク質(BMP)4および塩基性線維芽細胞増殖因子(bFGF)を含有する培養液中で培養し、さらにWnt阻害剤(IWP3)およびBMP4阻害剤(Dorsomorphin)およびTGFβ阻害剤(SB431542)を含む培養液中で培養し、その後VEGFおよびbFGFを含む培養液中で培養を行うことで、iPS細胞由来のヒト心筋細胞を得た。得られた細胞集団における心筋細胞の割合は50%~90%であった。 に お い て In the following examples, human iPS cells for clinical use established at Kyoto University iPS Cell Research Institute (CiRA) were used as pluripotent stem cells. Human iPS cells were maintained by a feeder-free method with reference to M. Nakagawa et al., Scientific Reports, 4: 3594 (2014). Then, human iPS cells were induced to differentiate into cardiomyocytes by referring to Miki et al., Cell Stem Cell 16, 16, 699-711, June 4, 2015 and WO 2014/185358 and WO 2017/038562 to induce embryoid bodies. I got Specifically, human iPS cells maintained and cultured in a culture solution containing no feeder cells were cultured on StemFit @ AK03 medium (Ajinomoto) containing 10 μM Y27632 (Wako Pure Chemical Industries) on EZ @ Sphere (Asahi Glass) for 1 day. Then, the obtained embryoid body was cultured in a culture solution containing activin A, bone morphogenetic protein (BMP) 4 and basic fibroblast growth factor (bFGF), and further inhibited with Wnt inhibitor (IWP3) and BMP4. The cells were cultured in a culture solution containing the agent (Dorsomorphin) and a TGFβ inhibitor (SB431542), and then cultured in a culture solution containing VEGF and bFGF to obtain iPS cell-derived human cardiomyocytes. The percentage of cardiomyocytes in the resulting cell population was between 50% and 90%.
例1.FBS含有培地とPL含有培地との比較
 上記で得られた、ヒトiPS細胞から分化誘導した心筋細胞を含む細胞集団を用い、シート化培養条件を検討した。DMEM/F12培地に20%FBSまたは5%ヒト血小板溶解物をそれぞれ加えたものをシート化媒体とした。血小板溶解物を加えたシート化媒体には、さらに細胞接着性成分としてラミニン(iMatrix-511)をそれぞれ0.1μg/mL、0.25μg/mLまたは0.5μg/mL加えた。また、シート化培養の1日目のみ、シート化媒体にRhoキナーゼ阻害剤Y27632を加えた。心筋細胞を含む細胞集団は、1.5×10個/cmの密度で温度応答性培養皿(UpCell(R)、セルシード)播種し、37℃、5%COの環境で3日間培養した。温度応答性培養皿は、培養液と同じもの(ただしY27632は含まない)を入れて、37℃で一晩インキュベートしてプレコーティングしたものを用いた。培養の後、培養皿から心筋細胞を含むシート状細胞培養物を剥離した 。
Example 1 Comparison between FBS-containing medium and PL-containing medium Using the cell population containing cardiomyocytes differentiated from human iPS cells obtained above, sheet-forming culture conditions were examined. A sheet medium was prepared by adding 20% FBS or 5% human platelet lysate to DMEM / F12 medium. To the sheeting medium to which the platelet lysate was added, laminin (iMatrix-511) was further added as a cell adhesive component at 0.1 μg / mL, 0.25 μg / mL or 0.5 μg / mL, respectively. Also, only on the first day of sheeting culture, the Rho kinase inhibitor Y27632 was added to the sheeting medium. Cell population comprising cardiomyocytes, temperature responsive culture dishes at a density of 1.5 × 10 6 cells / cm 2 (UpCell (R) , Cellseed) seeded, 37 ° C., 3 days of culture in the 5% CO 2 environment did. The temperature-responsive culture dish used was the same as the culture solution (excluding Y27632), and was incubated at 37 ° C. overnight and precoated. After the culture, the sheet-shaped cell culture containing cardiomyocytes was detached from the culture dish.
 結果を下表に示す。
Figure JPOXMLDOC01-appb-T000001
 FBSを入れたシート化媒体を用いた場合は、3日間シート化培養した時点では、拍動が観察されなかったり、また観察されても弱い拍動であったのに対し、ラミニンおよび血小板溶解物を入れたシート化媒体を用いたものは、いずれも3日間の培養で強い拍動が観察された。
The results are shown in the table below.
Figure JPOXMLDOC01-appb-T000001
In the case where the sheeting medium containing FBS was used, no pulsation was observed or weak pulsation was observed at the time of sheeting culture for 3 days, whereas laminin and platelet lysate were observed. In each of the cases using the sheet-form medium containing, strong pulsation was observed after culturing for 3 days.
例2.無血清培地とPL含有培地との比較
 例1と同様に、DMEM/F12培地にラミニン(iMatrix-511)を0.1μg/mL加えたものと、それにさらに5%の血小板溶解物を加えたものをそれぞれシート化媒体として、シート化培養を行った。
 結果を図1に示す。シート化培養2日目におけるシート化成功率を比較したところPL群では100%成功していたのに対し、無血清培地では50%であった。
Example 2. Similar to Comparative Example 1 between serum-free medium and PL-containing medium , DMEM / F12 medium supplemented with 0.1 μg / mL laminin (iMatrix-511), and further supplemented with 5% platelet lysate Were used as sheeting media to perform sheeting culture.
The results are shown in FIG. Comparing the sheeting success rate on the second day of sheeting culture, the PL group was 100% successful, whereas the serum-free medium was 50%.
 また、心筋細胞の拍動の強さを計測するため、セルモーションイメージングシステムSI8000(ソニー)を用いて拍動の様子を撮影し、Analyzer Softwareで拍動の加速度(acceleration)や拍動変形距離(contration deformation distance)を算出し、例1のFBS含有培地を用いた場合と例2のPL含有培地を用いた場合とで比較した。結果を図2に示す。PL含有培地を用いてシート状細胞培養物を形成した群は、FBS含有培地を用いた群と比べて、その拍動が強いことが確認された。無血清培地を用いた場合は、培養2日目までは拍動が観察されず、3日目にようやく拍動が観察されたのに対し、PL含有培地の場合は培養2日目において既に強い拍動が観察された。 In addition, in order to measure the intensity of the pulsation of the cardiomyocytes, the state of the pulsation was photographed using the cell motion imaging system SI8000 (Sony), and the pulsation acceleration (acceleration) and pulsation deformation distance ( contration deformation distance) was calculated and compared between the case where the FBS-containing medium of Example 1 was used and the case where the PL-containing medium of Example 2 was used. The results are shown in FIG. It was confirmed that the group in which the sheet-shaped cell culture was formed using the PL-containing medium had a stronger pulsation than the group using the FBS-containing medium. In the case of using the serum-free medium, no pulsation was observed until the second day of culture, and finally the pulsation was observed on the third day, whereas in the case of the PL-containing medium, the pulsation was already strong on the second day of culture. Beating was observed.
 これらのことから、PL含有培地を用いてシート化(移植片形成)をした場合は、無血清培地やFBS含有培地を用いた場合と比較して十分な細胞間ジャンクションの形成が達成されていることが予想される。そのためシート強度、心筋細胞の機能発現、成熟度などにおいて優れた移植片が形成できると考えられる。 From these facts, when the sheet is formed (graft formation) using the PL-containing medium, a sufficient intercellular junction is formed as compared with the case where the serum-free medium or the FBS-containing medium is used. It is expected that. Therefore, it is considered that an implant having excellent sheet strength, cardiomyocyte function expression, and maturity can be formed.
 本発明により、多能性幹細胞から分化誘導した細胞等を用いてシート状細胞培養物を形成する際に、高品質なシート状細胞培養物を得ることができる。特に臨床用に用いるゼノフリーなシート状細胞培養物の製造においても、高品質なシート状細胞培養物を簡便に形成することが可能となる。 According to the present invention, a high-quality sheet-shaped cell culture can be obtained when a sheet-shaped cell culture is formed using cells or the like that have been induced to differentiate from pluripotent stem cells. In particular, even in the production of a xeno-free sheet cell culture used for clinical use, a high-quality sheet cell culture can be easily formed.

Claims (8)

  1.  多能性幹細胞から分化誘導された細胞を含む移植片を製造する方法であって;
    (a)前記細胞を含む細胞集団を、培養基材上に播種する工程、および
    (b)播種した細胞集団を、血小板溶解物を含む移植片形成媒体で移植片形成培養する工程、
    を含む、前記方法。
    A method for producing a transplant containing cells induced to differentiate from pluripotent stem cells,
    (A) a step of inoculating a cell population containing the cells on a culture substrate, and (b) a step of explanting and culturing the inoculated cell population with an explant forming medium containing a platelet lysate;
    The above method, comprising:
  2.  多能性幹細胞が、iPS細胞である、請求項1に記載の方法。 方法 The method according to claim 1, wherein the pluripotent stem cells are iPS cells.
  3.  移植片が、シート状細胞培養物である、請求項1または2に記載の方法。 The method according to claim 1 or 2, wherein the graft is a sheet-shaped cell culture.
  4.  移植片形成媒体が、移植片に含まれる細胞種と異種の血清を含まない、請求項1~3のいずれか一項に記載の方法。 (4) The method according to any one of (1) to (3) above, wherein the graft forming medium does not contain serum different from the cell type contained in the graft.
  5.  移植片形成媒体が、さらに細胞接着性成分を含む、請求項1~4のいずれか一項に記載の方法。 (5) The method according to any one of (1) to (4), wherein the graft forming medium further comprises a cell adhesive component.
  6.  培養基材が、細胞接着性成分および/または血小板溶解物でコーティングされている、請求項1~5のいずれか一項に記載の方法。 The method according to any one of claims 1 to 5, wherein the culture substrate is coated with a cell adhesive component and / or a platelet lysate.
  7.  細胞が、心筋細胞である、請求項1~6のいずれか一項に記載の方法。 The method according to any one of claims 1 to 6, wherein the cell is a cardiomyocyte.
  8.  iPS細胞から分化誘導した心筋細胞を含む移植片において該心筋細胞の拍動開始を早める方法であって、前記心筋細胞を含む細胞集団を、血小板溶解物を含む移植片形成媒体で移植片形成培養することを含む、前記方法。 A method for accelerating the onset of pulsation of cardiomyocytes in a graft containing cardiomyocytes differentiated from iPS cells, wherein the cell population containing the cardiomyocytes is cultured in a graft-forming medium containing a platelet lysate. The above method, comprising:
PCT/JP2019/038192 2018-09-27 2019-09-27 Sheeting method for cardiomyocytes WO2020067439A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2020549444A JP7256818B2 (en) 2018-09-27 2019-09-27 Sheeting method of cardiomyocytes

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2018182443 2018-09-27
JP2018-182443 2018-09-27

Publications (1)

Publication Number Publication Date
WO2020067439A1 true WO2020067439A1 (en) 2020-04-02

Family

ID=69953538

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2019/038192 WO2020067439A1 (en) 2018-09-27 2019-09-27 Sheeting method for cardiomyocytes

Country Status (2)

Country Link
JP (1) JP7256818B2 (en)
WO (1) WO2020067439A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112272699A (en) * 2018-07-06 2021-01-26 株式会社迈傲锐治 Method for producing cell sheet, myocardial cell sheet, and kit for producing myocardial cell sheet
WO2022080455A1 (en) * 2020-10-14 2022-04-21 ダ・ヴィンチ ユニバーサル株式会社 Method for producing cardiac muscle stem/precursor cells and method for inhibiting myocardial fibrosis

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016076368A1 (en) * 2014-11-12 2016-05-19 テルモ株式会社 Myocardial cell sheet

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5710634B2 (en) * 2010-09-30 2015-04-30 国立大学法人 東京大学 Method for differentiating human-derived pluripotent stem cells
JP6948261B2 (en) * 2015-07-15 2021-10-13 国立大学法人大阪大学 Method for cryopreserving pluripotent stem cells or cardiomyocytes derived from adipose tissue or bone marrow-derived mesenchymal stem cells

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016076368A1 (en) * 2014-11-12 2016-05-19 テルモ株式会社 Myocardial cell sheet

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BURNOUF, THIERRY ET AL.: "Human platelet lysate: replacing fetal bovine serum as a gold standard for human cell propagation?", BIOMATERIALS, vol. 76, 2016, pages 371 - 387, XP029317322, DOI: 10.1016/j.biomaterials.2015.10.065 *
HAMANO, SAYURI ET AL.: "Extracellular Matrix from Periodontal Ligament Cells Could Induce the Differentiation of Induced Pluripotent Stem Cells to Periodontal Ligament Stem Cell -Like Cells", STEM CELLS AND DEVELOPMENT, vol. 27, no. 2, January 2018 (2018-01-01), pages 100 - 111, XP055701589, [retrieved on 20191202] *
SAPORITO, FRANCE SCA ET AL.: "Electrospun Gelatin- Chondroitin Sulfate Scaffolds Loaded with Platelet Lysate Promote Immature Cardiomyocyte Proliferation", POLYMERS, vol. 10, 21 February 2018 (2018-02-21), pages 1 - 20, XP55701574, [retrieved on 20191202] *
TIAN, BEIMIN ET AL.: "Human platelet lysate supports the formation of robust human periodontal ligament cell sheets", J. TISSUE ENG REGEN MED, vol. 12, 17 July 2017 (2017-07-17), pages 961 - 972, XP055701079, Retrieved from the Internet <URL:https://doi.org/10.1002/term.2511> [retrieved on 20191202] *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112272699A (en) * 2018-07-06 2021-01-26 株式会社迈傲锐治 Method for producing cell sheet, myocardial cell sheet, and kit for producing myocardial cell sheet
WO2022080455A1 (en) * 2020-10-14 2022-04-21 ダ・ヴィンチ ユニバーサル株式会社 Method for producing cardiac muscle stem/precursor cells and method for inhibiting myocardial fibrosis

Also Published As

Publication number Publication date
JPWO2020067439A1 (en) 2021-10-07
JP7256818B2 (en) 2023-04-12

Similar Documents

Publication Publication Date Title
Baldwin et al. In vitro pre-vascularisation of tissue-engineered constructs A co-culture perspective
JP6491187B2 (en) Use of microparticles and endothelial cells with decellularized organs and tissues
US20060153815A1 (en) Tissue engineering devices for the repair and regeneration of tissue
Chen et al. Biomaterial-assisted scalable cell production for cell therapy
Kuo et al. TATVHL peptide-grafted alginate/poly (γ-glutamic acid) scaffolds with inverted colloidal crystal topology for neuronal differentiation of iPS cells
JP2016528898A (en) Method for producing an artificial myocardium (EHM)
Matsuura et al. Toward the development of bioengineered human three-dimensional vascularized cardiac tissue using cell sheet technology
JP2009273444A (en) Cell-culturing supporter, method for producing the same and cell-culturing method using the supporter
CN107075443B (en) Three-dimensional cell culture system and cell culture method using the same
WO2020067439A1 (en) Sheeting method for cardiomyocytes
Poornejad et al. Current cell-based strategies for whole kidney regeneration
JP2014060991A (en) Method of culturing stem cells using inner lumen of porous hollow fiber
JP2023175787A (en) Method for forming pluripotent stem cell-derived cell into sheet
Gao et al. Recent progress in induced pluripotent stem cell-derived cardiac cell sheets for tissue engineering
Hudson et al. Development of myocardial constructs using modulus-matched acrylated polypropylene glycol triol substrate and different nonmyocyte cell populations
WO2021065984A1 (en) Method for forming sheet of cardiomyocytes
WO2020067443A1 (en) Somatic cell sheet-forming method
US20200109368A1 (en) Method for preparing differentiation-induced cells
WO2020067436A1 (en) Method for forming graft of pluripotent stem cell-derived cells
JP7471069B2 (en) Methods for enhancing graft activity
EP3739040B1 (en) Sheeting method for pluripotent stem cell-derived cells
Kiiskinen Co-culture of corneal epithelial cells and adipose stem cells-towards the use of hydrogels in ocular surface reconstruction
WO2021065989A1 (en) Method for increasing proportion of cd56+ cells
WO2021065976A1 (en) Method for enhancing activity in graft
Howard Review of Biomedical Applications of Cardiovascular Tissue Engineering

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19865383

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2020549444

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19865383

Country of ref document: EP

Kind code of ref document: A1